1
|
Suen CFDLW, Choy MC, Cruz PD. What to do when traditional rescue therapies fail in acute severe ulcerative colitis. Intest Res 2024; 22:397-413. [PMID: 38749658 PMCID: PMC11534448 DOI: 10.5217/ir.2024.00003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/15/2024] [Accepted: 03/22/2024] [Indexed: 06/12/2024] Open
Abstract
Acute severe ulcerative colitis (ASUC) is a medical emergency that affects approximately 25% of patients with ulcerative colitis at some point in time in their lives. Outcomes of ASUC are highly variable. Approximately 30% of patients do not respond to corticosteroids and up to 50% of patients do not respond to rescue therapy (infliximab or cyclosporin) and require emergency colectomy. Data are emerging on infliximab dosing strategies, use of cyclosporin as a bridge to slower acting biologic agents and Janus kinase inhibition as primary and sequential therapy. In this review, we outline contemporary approaches to clinical management of ASUC in the setting of failure to respond to traditional rescue therapies.
Collapse
Affiliation(s)
- Christopher F. D. Li Wai Suen
- Department of Gastroenterology, Austin Health, Melbourne, Australia
- Department of Medicine, Austin Academic Centre, University of Melbourne, Melbourne, Australia
| | - Matthew C. Choy
- Department of Gastroenterology, Austin Health, Melbourne, Australia
- Department of Medicine, Austin Academic Centre, University of Melbourne, Melbourne, Australia
| | - Peter De Cruz
- Department of Gastroenterology, Austin Health, Melbourne, Australia
- Department of Medicine, Austin Academic Centre, University of Melbourne, Melbourne, Australia
| |
Collapse
|
2
|
Shahraz A, Penney M, Candido J, Opoku‐Ansah G, Neubauer M, Eyles J, Ojo O, Liu N, Luheshi NM, Phipps A, Vishwanathan K. A mechanistic PK/PD model of AZD0171 (anti-LIF) to support Phase II dose selection. CPT Pharmacometrics Syst Pharmacol 2024; 13:1670-1681. [PMID: 39041713 PMCID: PMC11494920 DOI: 10.1002/psp4.13204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 06/04/2024] [Accepted: 07/02/2024] [Indexed: 07/24/2024] Open
Abstract
AZD0171 (INN: Falbikitug) is being developed as a humanized monoclonal antibody (mAb), immunoglobulin G subclass 1 (IgG1), which binds specifically to the immunosuppressive human cytokine leukemia inhibitory factor (LIF) and inhibits downstream signaling by blocking recruitment of glycoprotein 130 (gp130) to the LIF receptor (LIFR) subunit (gp190) and the phosphorylation of signal transducer and activator of transcription 3 (STAT3) and is intended to treat adult participants with advanced solid tumors. LIF is a pleiotropic cytokine (and a member of the IL-6 family of cytokines) involved in many physiological and pathological processes and is highly expressed in a subset of solid tumors, including non-small cell lung cancer (NSCLC), colon, ovarian, prostate, and pancreatic cancer. The aim of this work was to develop a mechanistic PK/PD model to investigate the effect of AZD0171 on tumor LIF levels, predict the level of downstream signaling complex (LIF:LIFR:gp130) inhibition, and examine the dose-response relationship to support dose selection for a Phase II clinical study. Modeling results show that tumor LIF is inhibited in a dose-dependent manner with >90% inhibition for 95% of patients at the Phase II clinical dose of 1500 mg Q2W.
Collapse
Affiliation(s)
- Azar Shahraz
- Clinical Pharmacology & Quantitative PharmacologyBioPharmaceuticals R&D, AstraZenecaWalthamMassachusettsUSA
| | - Mark Penney
- Early Oncology DMPK, Oncology R&D, AstraZenecaCambridgeUK
| | | | | | | | - Jim Eyles
- Oncology R&D, AstraZenecaCambridgeUK
| | | | | | | | - Alex Phipps
- Clinical Pharmacology & Quantitative PharmacologyBioPharmaceuticals R&D, AstraZenecaCambridgeUK
| | - Karthick Vishwanathan
- Clinical Pharmacology & Quantitative PharmacologyBioPharmaceuticals R&D, AstraZenecaWalthamMassachusettsUSA
| |
Collapse
|
3
|
Kumar M, Lanke S, Yadav A, Ette M, Mager DE, Shah DK. Inter-Antibody Variability in the Clinical Pharmacokinetics of Monoclonal Antibodies Characterized Using Population Physiologically Based Pharmacokinetic Modeling. Antibodies (Basel) 2024; 13:54. [PMID: 39051330 PMCID: PMC11270311 DOI: 10.3390/antib13030054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/27/2024] Open
Abstract
The objective of this work was to develop a population physiologically based pharmacokinetic (popPBPK) model to characterize the variability in the clinical PK of monoclonal antibodies (mAbs) following intravenous (IV) and subcutaneous (SC) administration. An extensive literature search was conducted and clinical PK data for FDA-approved as well as non-approved mAbs were collected. Training and validation datasets of 44 and 9 mAbs exhibiting linear pharmacokinetics were used for model development. The variability in antibody PK was captured by accounting for different rate constants of pinocytosis (CLup) and intracellular degradation (kdeg) for different mAbs. Typical values for CLup and kdeg and their respective inter-antibody variabilities (ωClup, ωKdeg) were estimated to be 0.32 L/h/L and 26.1 h-1 (73% and 46%). Varied absorption profiles following SC dosing were characterized by incorporating inter-antibody variability in local degradation (kSC) and rate of lymphatic uptake (S_Lu) of mAbs. Estimates for typical kSC and S_Lu values, and ωKsc,ωS_Lu, were found to be 0.0015 h-1 and 0.54 (193%, and 49%). FDA-approved mAbs showed less local degradation (0.0014 h-1 vs. 0.0038 h-1) compared with other clinically tested mAbs, whereas no substantial differences in physiological processes involved in disposition were observed. To evaluate the generalizability of estimated PK parameters and model validation, the final popPBPK model was used to simulate the range of expected PK for mAbs following SC administration of nine different mAbs that were not used for model-building purposes. The predicted PK of all nine mAbs was within the expected range specified a priori. Thus, the popPBPK model presented here may serve as a tool to predict the clinical PK of mAbs with linear disposition before administering them to humans. The model may also support preclinical-to-clinical translation and 'first-in-human' dose determination for mAbs.
Collapse
Affiliation(s)
- Mokshada Kumar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14214-8033, USA; (M.K.); (S.L.); (A.Y.); (M.E.); (D.E.M.)
| | - Sravani Lanke
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14214-8033, USA; (M.K.); (S.L.); (A.Y.); (M.E.); (D.E.M.)
| | - Alka Yadav
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14214-8033, USA; (M.K.); (S.L.); (A.Y.); (M.E.); (D.E.M.)
| | - Mfonabasi Ette
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14214-8033, USA; (M.K.); (S.L.); (A.Y.); (M.E.); (D.E.M.)
| | - Donald E. Mager
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14214-8033, USA; (M.K.); (S.L.); (A.Y.); (M.E.); (D.E.M.)
- Enhanced Pharmacodynamics, LLC, Buffalo, NY 14203, USA
| | - Dhaval K. Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14214-8033, USA; (M.K.); (S.L.); (A.Y.); (M.E.); (D.E.M.)
| |
Collapse
|
4
|
Perazzolo S. SAAM II: A general mathematical modeling rapid prototyping environment. CPT Pharmacometrics Syst Pharmacol 2024; 13:1088-1102. [PMID: 38863172 PMCID: PMC11247119 DOI: 10.1002/psp4.13181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/13/2024] Open
Abstract
Simulation Analysis and Modeling II (SAAM II) is a graphical modeling software used in life sciences for compartmental model analysis, particularly, but not exclusively, appreciated in pharmacokinetics (PK) and pharmacodynamics (PD), metabolism, and tracer modeling. Its intuitive "circles and arrows" visuals allow users to easily build, solve, and fit compartmental models without the need for coding. It is suitable for rapid prototyping of models for complex kinetic analysis or PK/PD problems, and in educating students and non-modelers. Although it is straightforward in design, SAAM II incorporates sophisticated algorithms programmed in C to address ordinary differential equations, deal with complex systems via forcing functions, conduct multivariable regression featuring the Bayesian maximum a posteriori, perform identifiability and sensitivity analyses, and offer reporting functionalities, all within a single package. After 26 years from the last SAAM II tutorial paper, we demonstrate here SAAM II's updated applicability to current life sciences challenges. We review its features and present four contemporary case studies, including examples in target-mediated PK/PD, CAR-T-cell therapy, viral dynamics, and transmission models in epidemiology. Through such examples, we demonstrate that SAAM II provides a suitable interface for rapid model selection and prototyping. By enabling the fast creation of detailed mathematical models, SAAM II addresses a unique requirement within the mathematical modeling community.
Collapse
Affiliation(s)
- Simone Perazzolo
- Nanomath LLC, Spokane, Washington, USA
- University of Washington, Seattle, Washington, USA
| |
Collapse
|
5
|
Rubahamya B, Dong S, Thurber GM. Clinical translation of antibody drug conjugate dosing in solid tumors from preclinical mouse data. SCIENCE ADVANCES 2024; 10:eadk1894. [PMID: 38820153 PMCID: PMC11141632 DOI: 10.1126/sciadv.adk1894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 04/29/2024] [Indexed: 06/02/2024]
Abstract
Antibody drug conjugates (ADCs) have made impressive strides in the clinic in recent years with 11 Food and Drug Administration approvals, including 6 for the treatment of patients with solid tumors. Despite this success, the development of new agents remains challenging with a high failure rate in the clinic. Here, we show that current approved ADCs for the treatment of patients with solid tumors can all show substantial efficacy in some mouse models when administered at a similar weight-based [milligrams per kilogram (mg/kg)] dosing in mice that is tolerated in the clinic. Mechanistically, equivalent mg/kg dosing results in a similar drug concentration in the tumor and a similar tissue penetration into the tumor due to the unique delivery features of ADCs. Combined with computational approaches, which can account for the complex distribution within the tumor microenvironment, these scaling concepts may aid in the evaluation of new agents and help design therapeutics with maximum clinical efficacy.
Collapse
Affiliation(s)
- Baron Rubahamya
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shujun Dong
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Greg M. Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
6
|
Asar M, Newton-Northup J, Soendergaard M. Improving Pharmacokinetics of Peptides Using Phage Display. Viruses 2024; 16:570. [PMID: 38675913 PMCID: PMC11055145 DOI: 10.3390/v16040570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Phage display is a versatile method often used in the discovery of peptides that targets disease-related biomarkers. A major advantage of this technology is the ease and cost efficiency of affinity selection, also known as biopanning, to identify novel peptides. While it is relatively straightforward to identify peptides with optimal binding affinity, the pharmacokinetics of the selected peptides often prove to be suboptimal. Therefore, careful consideration of the experimental conditions, including the choice of using in vitro, in situ, or in vivo affinity selections, is essential in generating peptides with high affinity and specificity that also demonstrate desirable pharmacokinetics. Specifically, in vivo biopanning, or the combination of in vitro, in situ, and in vivo affinity selections, has been proven to influence the biodistribution and clearance of peptides and peptide-conjugated nanoparticles. Additionally, the marked difference in properties between peptides and nanoparticles must be considered. While peptide biodistribution depends primarily on physiochemical properties and can be modified by amino acid modifications, the size and shape of nanoparticles also affect both absorption and distribution. Thus, optimization of the desired pharmacokinetic properties should be an important consideration in biopanning strategies to enable the selection of peptides and peptide-conjugated nanoparticles that effectively target biomarkers in vivo.
Collapse
Affiliation(s)
- Mallika Asar
- College of Osteopathic Medicine, Kansas City University, Kansas City, MO 64106, USA;
| | | | - Mette Soendergaard
- Cell Origins LLC, 1601 South Providence Road Columbia, Columbia, MO 65203, USA;
- Department of Chemistry, Western Illinois University, Macomb, IL 61455, USA
| |
Collapse
|
7
|
Ji Y, Sy SKB. Utility and impact of quantitative pharmacology on dose selection and clinical development of immuno-oncology therapy. Cancer Chemother Pharmacol 2024; 93:273-293. [PMID: 38430307 DOI: 10.1007/s00280-024-04643-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/23/2024] [Indexed: 03/03/2024]
Abstract
Immuno-oncology (IO) therapies have changed the cancer treatment landscape. Immune checkpoint inhibitors (ICIs) have improved overall survival in 20-40% of patients with malignancies that were previously refractory. Due to the uniqueness in biology, modalities and patient responses, drug development strategies for IO differed from that traditionally used for cytotoxic and target therapies in oncology, and quantitative pharmacology utilizing modeling approach can be applied in all phases of the development process. In this review, we used case studies to showcase how various modeling methodologies were applied from translational science and dose selection through to label change, using examples that included anti-programmed-death-1 (anti-PD-1), anti-programmed-death ligand-1 (anti-PD-L1), anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4), and anti-glucocorticoid-induced tumor necrosis factor receptor-related protein (anti-GITR) antibodies. How these approaches were utilized to support phase I-III dose selection, the design of phase III trials, and regulatory decisions on label change are discussed to illustrate development strategies. Model-based quantitative approaches have positively impacted IO drug development, and a better understanding of the biology and exposure-response relationship may benefit the development and optimization of new IO therapies.
Collapse
Affiliation(s)
- Yan Ji
- Novartis Pharmaceuticals Corporation, 1 Health Plaza, East Hanover, NJ, 07936, USA.
| | - Sherwin K B Sy
- Novartis Pharmaceuticals Corporation, 1 Health Plaza, East Hanover, NJ, 07936, USA.
| |
Collapse
|
8
|
GomezMancilla B, Meriggioli MN, Genge A, Roubenoff R, Espié P, Dupuy C, Hartmann N, Pezous N, Kinhikar A, Tichy M, Dionne A, Vissing J, Andersen H, Schoser B, Meisel A, Jordan B, Devlikamova F, Poverennova I, Stuchevskaya F, Lin TS, Rush JS, Gergely P. Efficacy and safety of iscalimab, a novel anti-CD40 monoclonal antibody, in moderate-to-severe myasthenia gravis: A phase 2 randomized study. J Clin Neurosci 2024; 119:76-84. [PMID: 37988976 DOI: 10.1016/j.jocn.2023.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/19/2023] [Accepted: 11/07/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND Increased morbidity in many patients with myasthenia gravis (MG) on long-term immunosuppression highlights the need for improved treatments. The aim of this study is to investigate the safety and efficacy of iscalimab (CFZ533), a fully human anti-CD40 monoclonal antibody, in patients with moderate-to-severe MG receiving standard-of-care (SoC) therapies. METHODS In this double-blind, placebo-controlled phase 2 study, symptomatic patients (n = 44) despite SoC were randomized 1:1 to receive intravenous iscalimab (10 mg/kg; n = 22) or placebo (n = 22) every 4 weeks for 6 doses in total. Patients were followed up for 6 months after the last dose. The total duration of the study was 52 weeks. RESULTS In total, 34 of 44 patients (77.3 %) completed the study. The primary endpoint, Quantitative MG score, did not change significantly between baseline and week 25 for iscalimab (median [90 % CI], -4.07 [-5.67, -2.47]) versus placebo (-2.93 [-4.53, -1.33]); however, non-thymectomized patients (n = 29) showed more favorable results (iscalimab, -4.35 [-6.07, -2.64] vs placebo, -2.26 [-4.16, -0.36]). A statistically significant difference between iscalimab and placebo groups was observed in MG Composite score (adjusted mean change: -4.19 [-6.67, -1.72]; p = 0.007) at week 13, and MG-Activities of Daily Living score (-1.93 [-3.24, -0.62]; p = 0.018) at week 21. Adverse events were comparable between the iscalimab (91 %) and placebo (96 %) groups. CONCLUSION Iscalimab showed favorable safety and improvements compared with placebo in non-thymectomized patients with moderate-to-severe MG. It did not show any protective effect in patients with moderate-to-severe MG.
Collapse
Affiliation(s)
| | | | - Angela Genge
- Montreal Neurological Institute and Hospital, Montreal, Canada
| | | | - Pascal Espié
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Cyrielle Dupuy
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Nicole Hartmann
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Nicole Pezous
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Arvind Kinhikar
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Mia Tichy
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - John Vissing
- Rigshospitalet, University of Copenhagen, Denmark
| | | | - Benedikt Schoser
- Friedrich-Baur-Institute, Dep. of Neurology, LMU Klinikum Muenchen, Muenchen, Germany
| | - Andreas Meisel
- Charité Universitätsmedizin Berlin, Department of Neurology, Germany
| | - Berit Jordan
- Department of Neurology, University Hospital of Halle, Halle, Germany; epartment of Neurology, University Hospital of Heidelberg, Heidelberg, Germany
| | | | - Irina Poverennova
- Samara Regional Clinical Hospital named after M.I.Kalinin, Samara, Russia
| | | | - Thy-Sheng Lin
- National Cheng Kung University Hospital, Tainan, Taiwan
| | - James S Rush
- Novartis Institutes for BioMedical Research, Basel, Switzerland; Kling Biotherapeuetics BV, Amsterdam, the Netherlands
| | - Peter Gergely
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| |
Collapse
|
9
|
Augustin D, Lambert B, Robinson M, Wang K, Gavaghan D. Simulating clinical trials for model-informed precision dosing: using warfarin treatment as a use case. Front Pharmacol 2023; 14:1270443. [PMID: 37927586 PMCID: PMC10621790 DOI: 10.3389/fphar.2023.1270443] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Treatment response variability across patients is a common phenomenon in clinical practice. For many drugs this inter-individual variability does not require much (if any) individualisation of dosing strategies. However, for some drugs, including chemotherapies and some monoclonal antibody treatments, individualisation of dosages are needed to avoid harmful adverse events. Model-informed precision dosing (MIPD) is an emerging approach to guide the individualisation of dosing regimens of otherwise difficult-to-administer drugs. Several MIPD approaches have been suggested to predict dosing strategies, including regression, reinforcement learning (RL) and pharmacokinetic and pharmacodynamic (PKPD) modelling. A unified framework to study the strengths and limitations of these approaches is missing. We develop a framework to simulate clinical MIPD trials, providing a cost and time efficient way to test different MIPD approaches. Central for our framework is a clinical trial model that emulates the complexities in clinical practice that challenge successful treatment individualisation. We demonstrate this framework using warfarin treatment as a use case and investigate three popular MIPD methods: 1. Neural network regression; 2. Deep RL; and 3. PKPD modelling. We find that the PKPD model individualises warfarin dosing regimens with the highest success rate and the highest efficiency: 75.1% of the individuals display INRs inside the therapeutic range at the end of the simulated trial; and the median time in the therapeutic range (TTR) is 74%. In comparison, the regression model and the deep RL model have success rates of 47.0% and 65.8%, and median TTRs of 45% and 68%. We also find that the MIPD models can attain different degrees of individualisation: the Regression model individualises dosing regimens up to variability explained by covariates; the Deep RL model and the PKPD model individualise dosing regimens accounting also for additional variation using monitoring data. However, the Deep RL model focusses on control of the treatment response, while the PKPD model uses the data also to further the individualisation of predictions.
Collapse
Affiliation(s)
- David Augustin
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Ben Lambert
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, United Kingdom
| | - Martin Robinson
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Ken Wang
- Research and Early Development, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - David Gavaghan
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
10
|
Lobet S, Paintaud G, Azzopardi N, Passot C, Caulet M, Chautard R, Desvignes C, Capitain O, Tougeron D, Lecomte T, Ternant D. Relationship Between Cetuximab Target-Mediated Pharmacokinetics and Progression-Free Survival in Metastatic Colorectal Cancer Patients. Clin Pharmacokinet 2023; 62:1263-1274. [PMID: 37442917 DOI: 10.1007/s40262-023-01270-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2023] [Indexed: 07/15/2023]
Abstract
BACKGROUND AND OBJECTIVE Cetuximab, an anti-epidermal growth factor receptor (EGFR) monoclonal immunoglobulin (Ig)G1 antibody, has been approved for the treatment of metastatic colorectal cancer (mCRC). The influence of target-antigen on cetuximab pharmacokinetics has never been investigated using target-mediated drug disposition (TMDD) modelling. This study aimed to investigate the relationship between cetuximab concentrations, target kinetics and progression-free survival (PFS). METHODS In this ancillary study (NCT00559741), 91 patients with mCRC treated with cetuximab were assessed. Influence of target levels on cetuximab pharmacokinetics was described using TMDD modelling. The relationship between cetuximab concentrations, target kinetics and time-to-progression (TTP) was described using a joint pharmacokinetic-TTP model, where unbound target levels were assumed to influence hazard of progression by an Emax model. Mitigation strategies of concentration-response relationship, i.e., time-varying endogenous clearance and mutual influences of clearance and time-to-progression were investigated. RESULTS Cetuximab concentration-time data were satisfactorily described using the TMDD model with quasi-steady-state approximation and time-varying endogenous clearance. Estimated target parameters were baseline target levels (R0 = 43 nM), and complex elimination rate constant (kint = 0.95 day-1). Estimated time-varying clearance parameters were time-invariant component of CL (CL0= 0.38 L/day-1), time-variant component of CL (CL1= 0.058 L/day-1) and first-order rate of CL1 decreasing over time (kdes = 0.049 day-1). Part of concentration-TTP was TTP-driven, where clearance and TTP were inversely correlated. In addition, increased target occupancy was associated with increased TTP. CONCLUSION This is the first study describing the complex relationship between cetuximab target-mediated pharmacokinetics and PFS in mCRC patients using a joint PK-time-to-progression model. Further studies are needed to provide a more in-depth description of this relationship.
Collapse
Affiliation(s)
- Sarah Lobet
- Inserm UMR 1069, Nutrition Croissance et Cancer (N2C), Tours University, Tours, France
| | - Gilles Paintaud
- EA4245 Transplantation, Immunologie, Inflammation (T2i), Tours University, Tours, France
- Centre Pilote de suivi Biologique des traitements par Anticorps (CePiBAc), Tours University Hospital, Tours, France
- Pharmacology-Toxicology Department, Tours University Hospital, Tours, France
| | | | - Christophe Passot
- Oncopharmacology-Pharmacogenetics Department INSERM U892, Institut de Cancérologie de l'Ouest site Paul Papin, Angers, France
| | - Morgane Caulet
- Gastroenterology and Digestive oncology Department, Tours University Hospital, Tours, France
| | - Romain Chautard
- Inserm UMR 1069, Nutrition Croissance et Cancer (N2C), Tours University, Tours, France
- Gastroenterology and Digestive oncology Department, Tours University Hospital, Tours, France
| | - Céline Desvignes
- EA4245 Transplantation, Immunologie, Inflammation (T2i), Tours University, Tours, France
- Centre Pilote de suivi Biologique des traitements par Anticorps (CePiBAc), Tours University Hospital, Tours, France
| | - Olivier Capitain
- Oncopharmacology-Pharmacogenetics Department INSERM U892, Institut de Cancérologie de l'Ouest site Paul Papin, Angers, France
| | - David Tougeron
- Gastroenterology Department, Poitiers University Hospital, Poitiers, France
- PRoDiCeT, Poitiers University, Poitiers, France
| | - Thierry Lecomte
- Inserm UMR 1069, Nutrition Croissance et Cancer (N2C), Tours University, Tours, France
- Gastroenterology and Digestive oncology Department, Tours University Hospital, Tours, France
| | - David Ternant
- EA4245 Transplantation, Immunologie, Inflammation (T2i), Tours University, Tours, France.
- Centre Pilote de suivi Biologique des traitements par Anticorps (CePiBAc), Tours University Hospital, Tours, France.
- Pharmacology-Toxicology Department, Tours University Hospital, Tours, France.
| |
Collapse
|
11
|
Li X, Dosne AG, Pérez Ruixo C, Perez Ruixo JJ. Pharmacodynamic-Mediated Drug Disposition (PDMDD) Model of Daratumumab Monotherapy in Patients with Multiple Myeloma. Clin Pharmacokinet 2023; 62:761-777. [PMID: 37022569 DOI: 10.1007/s40262-023-01232-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND AND OBJECTIVE We aimed to quantify the daratumumab concentration- and CD38 dynamics-dependent pharmacokinetics using a pharmacodynamic mediated disposition model (PDMDD) in patients with multiple myeloma (MMY) following daratumumab IV or SC monotherapy. Daratumumab, a human IgG monoclonal antibody targeting CD38 with a direct on-tumor and immunomodulatory mechanism of action, has been approved to treat patients with multiple myeloma (MM). METHODS In total, 7788 daratumumab plasma samples from 850 patients with diagnosis of MMY were used. The serum concentration-time data of daratumumab were analysed using nonlinear mixed-effects modeling with NONMEM®. The PDMDD with quasi steady-state approximation (QSS) was compared to the previously developed Michaelis-Menten (MM) approximation with respect to the parameter estimates, the goodness-of-fit plots and prediction-corrected visual predictive check, as well as model-based simulations. The effect of patients' covariates on daratumumab pharmacokinetics was also investigated. RESULTS The QSS approximation characterized the concentration- and CD38 dynamics-dependency of daratumumab pharmacokinetics within the doses ranging from 0.1 to 24 mg/kg after IV administration and 1200 and 1800 mg after SC administration in patients with MMY, mechanistically describing the binding of daratumumab and CD38, the internalization of the daratumumab-CD38 complex and the CD38 turnover. Compared to the previously developed MM approximation, the MM approximation with the non-constant total target and dose-correction provided substantial improvement of the model fit, but it was still not as good as the QSS approximation. The effect of the previously identified covariates as well as the newly identified covariate (baseline M protein) on daratumumab pharmacokinetics was confirmed, but the magnitude of the effect was deemed not clinically relevant. CONCLUSIONS Accounting for the CD38 turnover and its binding capacity to daratumumab, the QSS approximation provided a mechanistic interpretation of daratumumab PK parameters and consequently well described the concentration- and CD38 dynamics-dependency of daratumumab pharmacokinetics. CLINICAL STUDIES INCLUDED IN THE ANALYSIS WERE REGISTERED WITH THE NCT NUMBER BELOW AT HTTP://WWW. CLINICALTRIALS GOV : MMY1002 (ClinicalTrials.gov: NCT02116569), MMY1003 (NCT02852837), MMY1004 (NCT02519452), MMY1008 (NCT03242889), GEN501 (NCT00574288), MMY2002 (NCT01985126), MMY3012 (NCT03277105).
Collapse
Affiliation(s)
- Xia Li
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development LLC, Beerse, Belgium.
| | - Anne-Gaëlle Dosne
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development LLC, Beerse, Belgium
| | - Carlos Pérez Ruixo
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development LLC, Beerse, Belgium
| | - Juan Jose Perez Ruixo
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development LLC, Beerse, Belgium
| |
Collapse
|
12
|
Guo G, You X, Wu W, Chen J, Ke M, Lin R, Huang P, Lin C. Physiologically-Based Pharmacokinetic Modeling of Omalizumab to Predict the Pharmacokinetics and Pharmacodynamics in Pediatric Patients. Clin Pharmacol Ther 2023; 113:724-734. [PMID: 36495063 DOI: 10.1002/cpt.2815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Omalizumab is widely used in clinical practice; however, knowledge gaps in the dosage of omalizumab for children aged 2-6 years with moderate-to-severe persistent allergic asthma have been identified. The aim of this study was to explore dosing regimens for moderately-to-severely allergic pediatric patients aged 2-6 years. The physiologically-based pharmacokinetic (PBPK) model of omalizumab was developed and verified in adult patients, extrapolated to pediatric patients, and simulated for omalizumab by adding two observation chambers (free IgE and total IgE). The simulation results showed that the fold errors of the predicted and observed values of the area under the curve (AUC) and peak plasma concentration (Cmax ) were between 0.5 and 2.0, and the average folding error and the absolute average folding error values for all concentration-time data points were 1.09 and 1.48, respectively. The PBPK model combined with pharmacokinetic/pharmacodynamic analysis of omalizumab demonstrated that both the model-derived dose and the original dose could control the average free IgE of 2-6-year-old children with moderate-to-severe allergic asthma below 25 ng/mL, and some of the model-derived doses were lower. This conclusion provides a basis for the selection of dosage in clinical practice reference.
Collapse
Affiliation(s)
- Guimu Guo
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiang You
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Wanhong Wu
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jiarui Chen
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Meng Ke
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Rongfang Lin
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Pinfang Huang
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Cuihong Lin
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
13
|
Sy SKB, Tanaka C, Grosch K. Population Pharmacokinetics and Pharmacodynamics of Crizanlizumab in Healthy Subjects and Patients with Sickle Cell Disease. Clin Pharmacokinet 2023; 62:249-266. [PMID: 36529836 DOI: 10.1007/s40262-022-01193-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND OBJECTIVES Crizanlizumab is a humanized monoclonal antibody against P-selectin for the prevention of vaso-occlusive crises in sickle cell disease (SCD). The objective of this study was to investigate crizanlizumab population pharmacokinetics (PK) and pharmacodynamics (PD), as well as influential covariates. METHODS A population PK model for crizanlizumab was developed from healthy volunteer and SCD patient data, using a two-compartment intravenous infusion model utilizing a target-mediated drug disposition (TMDD) approach. The relationship between crizanlizumab concentration and ex vivo P-selectin inhibition was fitted to a non-linear sigmoidal Emax model. Covariate selection was performed in a stepwise manner. RESULTS Crizanlizumab exhibits nonlinear pharmacokinetics in the wide dose range of 0.2-8 mg/kg body weight. The population pharmacokinetic base model incorporated body weight as covariate in the form of allometric scaling wherein the exponents were fixed to 0.8. SCD patients had higher baseline soluble P-selectin concentration, resulting in a higher estimated initial target concentration. The typical individual in the model is a 70 kg SCD patient with normal renal function and a baseline albumin of 43 g/L; CL was 0.012 L/h while Vss was 5.2 L. For the population PD model, none of the identified additional factors beyond PD assay and covariates, such as body weight at baseline nor patient type differences, led to relevant differences in P-selectin % inhibition. CONCLUSIONS Renal and hepatic impairments, concomitant hydroxyurea usage, and presence of anti-drug antibody are not expected to impact the exposure of crizanlizumab. The model allows for extrapolating the PK of crizanlizumab to pediatric population and evaluation of alternative regimens and route of administration. TRIAL REGISTRATION NUMBER [DATE OF REGISTRATION]: SUSTAIN (CSEG101A2201 Phase 2), ClinicalTrials.gov identifier: NCT01895361 [10 July 2013]; CSEG101A2202 (Phase 2), ClinicalTrials.gov identifier: NCT03264989 [29 August 2017].
Collapse
Affiliation(s)
- Sherwin K B Sy
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936-1080, USA.
| | - Chiaki Tanaka
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936-1080, USA
| | - Kai Grosch
- Novartis Pharma AG, Forum 1, Novartis Campus, 4056, Basel, Switzerland
| |
Collapse
|
14
|
Menezes B, Khera E, Calopiz M, Smith MD, Ganno ML, Cilliers C, Abu-Yousif AO, Linderman JJ, Thurber GM. Pharmacokinetics and Pharmacodynamics of TAK-164 Antibody Drug Conjugate Coadministered with Unconjugated Antibody. AAPS J 2022; 24:107. [PMID: 36207468 PMCID: PMC10754641 DOI: 10.1208/s12248-022-00756-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/21/2022] [Indexed: 11/24/2022] Open
Abstract
The development of new antibody-drug conjugates (ADCs) has led to the approval of 7 ADCs by the FDA in 4 years. Given the impact of intratumoral distribution on efficacy of these therapeutics, coadministration of unconjugated antibody with ADC has been shown to improve distribution and efficacy of several ADCs in high and moderately expressed tumor target systems by increasing tissue penetration. However, the benefit of coadministration in low expression systems is less clear. TAK-164, an ADC composed of an anti-GCC antibody (5F9) conjugated to a DGN549 payload, has demonstrated heterogeneous distribution and bystander killing. Here, we evaluated the impact of 5F9 coadministration on distribution and efficacy of TAK-164 in a primary human tumor xenograft mouse model. Coadministration was found to improve the distribution of TAK-164 within the tumor, but it had no significant impact (increase or decrease) on efficacy. Experimental and computational evidence indicates that this was not a result of tumor saturation, increased binding to perivascular cells, or compensatory bystander effects. Rather, the cellular potency of DGN549 was matched with the single-cell uptake of TAK-164 making its IC50 close to its equilibrium binding affinity (KD), and as such, coadministration dilutes total DGN549 in cells below the maximum cytotoxic concentration, thereby offsetting an increased number of targeted cells with decreased ability to kill each cell. These results provide new insights on matching payload potency to ADC delivery to help identify when increasing tumor penetration is beneficial for improving ADC efficacy and demonstrate how mechanistic simulations can be leveraged to design clinically effective ADCs.
Collapse
Affiliation(s)
- Bruna Menezes
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd, Ann Arbor, Michigan, 48109, USA
| | - Eshita Khera
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd, Ann Arbor, Michigan, 48109, USA
| | - Melissa Calopiz
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd, Ann Arbor, Michigan, 48109, USA
| | - Michael D Smith
- Takeda Development Center Americas-Inc. TDCA, Oncology, Lexington, Massachussetts, USA
| | - Michelle L Ganno
- Takeda Development Center Americas-Inc. TDCA, Oncology, Lexington, Massachussetts, USA
| | - Cornelius Cilliers
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd, Ann Arbor, Michigan, 48109, USA
| | - Adnan O Abu-Yousif
- Takeda Development Center Americas-Inc. TDCA, Oncology, Lexington, Massachussetts, USA
| | - Jennifer J Linderman
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd, Ann Arbor, Michigan, 48109, USA
- Department of Biomedical Engineering, University of Michigan, 2800 Plymouth Rd, Ann Arbor, Michigan, 48109, USA
| | - Greg M Thurber
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd, Ann Arbor, Michigan, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, 2800 Plymouth Rd, Ann Arbor, Michigan, 48109, USA.
| |
Collapse
|
15
|
A long-acting C-natriuretic peptide for achondroplasia. Proc Natl Acad Sci U S A 2022; 119:e2201067119. [PMID: 35858423 PMCID: PMC9335275 DOI: 10.1073/pnas.2201067119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The C-natriuretic peptide (CNP) analog vosoritide has recently been approved for treatment of achondroplasia in children. However, the regimen requires daily subcutaneous injections in pediatric patients over multiple years. The present work sought to develop a long-acting CNP that would provide efficacy equal to or greater than that of vosoritide but require less frequent injections. We used a technology for half-life extension, whereby a drug is attached to tetra-polyethylene glycol hydrogels (tetra-PEG) by β-eliminative linkers that cleave at predetermined rates. These hydrogels-fabricated as uniform ∼60-μm microspheres-are injected subcutaneously, where they serve as a stationary depot to slowly release the drug into the systemic circulation. We prepared a highly active, stable CNP analog-[Gln6,14]CNP-38-composed of the 38 C-terminal amino acids of human CNP-53 containing Asn to Gln substitutions to preclude degradative deamidation. Two microsphere [Gln6,14]CNP-38 conjugates were prepared, with release rates designed to allow once-weekly and once-monthly administration. After subcutaneous injection of the conjugates in mice, [Gln6,14]CNP-38 was slowly released into the systemic circulation and showed biphasic elimination pharmacokinetics with terminal half-lives of ∼200 and ∼600 h. Both preparations increased growth of mice comparable to or exceeding that produced by daily vosoritide. Simulations of the pharmacokinetics in humans indicated that plasma [Gln6,14]CNP-38 levels should be maintained within a therapeutic window over weekly, biweekly, and likely, monthly dosing intervals. Compared with vosoritide, which requires ∼30 injections per month, microsphere [Gln6,14]CNP-38 conjugates-especially the biweekly and monthly dosing-could provide an alternative that would be well accepted by physicians, patients, and patient caregivers.
Collapse
|
16
|
Haraya K, Tsutsui H, Komori Y, Tachibana T. Recent Advances in Translational Pharmacokinetics and Pharmacodynamics Prediction of Therapeutic Antibodies Using Modeling and Simulation. Pharmaceuticals (Basel) 2022; 15:ph15050508. [PMID: 35631335 PMCID: PMC9145563 DOI: 10.3390/ph15050508] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 02/05/2023] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) have been a promising therapeutic approach for several diseases and a wide variety of mAbs are being evaluated in clinical trials. To accelerate clinical development and improve the probability of success, pharmacokinetics and pharmacodynamics (PKPD) in humans must be predicted before clinical trials can begin. Traditionally, empirical-approach-based PKPD prediction has been applied for a long time. Recently, modeling and simulation (M&S) methods have also become valuable for quantitatively predicting PKPD in humans. Although several models (e.g., the compartment model, Michaelis–Menten model, target-mediated drug disposition model, and physiologically based pharmacokinetic model) have been established and used to predict the PKPD of mAbs in humans, more complex mechanistic models, such as the quantitative systemics pharmacology model, have been recently developed. This review summarizes the recent advances and future direction of M&S-based approaches to the quantitative prediction of human PKPD for mAbs.
Collapse
Affiliation(s)
- Kenta Haraya
- Discovery Biologics Department, Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan;
- Correspondence:
| | - Haruka Tsutsui
- Discovery Biologics Department, Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan;
| | - Yasunori Komori
- Pharmaceutical Science Department, Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan; (Y.K.); (T.T.)
| | - Tatsuhiko Tachibana
- Pharmaceutical Science Department, Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan; (Y.K.); (T.T.)
| |
Collapse
|
17
|
Peletier LA, Gabrielsson J. Impact of enzyme turnover on the dynamics of the Michaelis–Menten model. Math Biosci 2022; 346:108795. [DOI: 10.1016/j.mbs.2022.108795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 11/29/2022]
|
18
|
Saghari M, Gal P, Gilbert S, Yateman M, Porter‐Brown B, Brennan N, Quaratino S, Wilson R, Grievink HW, Klaassen ES, Bergmann KR, Burggraaf J, Doorn MB, Powell J, Moerland M, Rissmann R. OX40L Inhibition Suppresses KLH‐driven Immune Responses in Healthy Volunteers: A Randomized Controlled Trial Demonstrating Proof‐of‐Pharmacology for KY1005. Clin Pharmacol Ther 2022; 111:1121-1132. [PMID: 35092305 PMCID: PMC9314635 DOI: 10.1002/cpt.2539] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/20/2022] [Indexed: 11/24/2022]
Abstract
The safety, tolerability, immunogenicity, and pharmacokinetic (PK) profile of an anti‐OX40L monoclonal antibody (KY1005, currently amlitelimab) were evaluated. Pharmacodynamic (PD) effects were explored using keyhole limpet hemocyanin (KLH) and tetanus toxoid (TT) immunizations. Sixty‐four healthy male subjects (26.5 ± 6.0 years) were randomized to single doses of 0.006, 0.018, or 0.05 mg/kg, or multiple doses of 0.15, 0.45, 1.35, 4, or 12 mg/kg KY1005, or placebo (6:2). Serum KY1005 concentrations were measured. Antibody responses upon KLH and TT immunizations and skin response upon intradermal KLH administration were performed. PD data were analyzed using repeated measures analysis of covariances (ANCOVAs) and post hoc exposure‐response modeling. No serious adverse events occurred and all adverse events were temporary and of mild or moderate severity. A nonlinear increase in mean serum KY1005 concentrations was observed (median time to maximum concentration (Tmax) ~ 4 hours, geometric mean terminal half‐life (t½) ~ 24 days). Cutaneous blood perfusion (estimated difference (ED) −13.4 arbitrary unit (AU), 95% confidence interval (CI) −23.0 AU to −3.8 AU) and erythema quantified as average redness (ED −0.23 AU, 95% CI −0.35 AU to −0.11 AU) decreased after KY1005 treatment at doses of 0.45 mg/kg and above. Exposure‐response analysis displayed a statistically significant treatment effect on anti‐KLH antibody titers (IgG maximum effect (Emax) −0.58 AU, 95% CI −1.10 AU to −0.06 AU) and skin response (erythema Emax −0.20 AU, 95% CI −0.29 AU to −0.11 AU). Administration of KY1005 demonstrated an acceptable safety and tolerability profile and PK analyses displayed a nonlinear profile of KY1005. Despite the observed variability, skin challenge response after KY1005 treatment indicated pharmacological activity of KY1005. Therefore, KY1005 shows potential as a novel pharmacological treatment in immune‐mediated disorders.
Collapse
Affiliation(s)
- Mahdi Saghari
- Centre for Human Drug Research Leiden the Netherlands
- Leiden University Medical Centre Leiden the Netherlands
| | - Pim Gal
- Centre for Human Drug Research Leiden the Netherlands
- Leiden University Medical Centre Leiden the Netherlands
| | | | | | | | | | | | | | - Hendrika W. Grievink
- Centre for Human Drug Research Leiden the Netherlands
- Leiden Academic Centre for Drug Research Leiden the Netherlands
| | | | | | - Jacobus Burggraaf
- Centre for Human Drug Research Leiden the Netherlands
- Leiden University Medical Centre Leiden the Netherlands
- Leiden Academic Centre for Drug Research Leiden the Netherlands
| | - Martijn B.A. Doorn
- Department of Dermatology Erasmus Medical Centre Rotterdam the Netherlands
| | | | - Matthijs Moerland
- Centre for Human Drug Research Leiden the Netherlands
- Leiden University Medical Centre Leiden the Netherlands
| | - Robert Rissmann
- Centre for Human Drug Research Leiden the Netherlands
- Leiden University Medical Centre Leiden the Netherlands
- Leiden Academic Centre for Drug Research Leiden the Netherlands
| |
Collapse
|
19
|
Chang HY, Wu S, Chowdhury EA, Shah DK. Towards a translational physiologically-based pharmacokinetic (PBPK) model for receptor-mediated transcytosis of anti-transferrin receptor monoclonal antibodies in the central nervous system. J Pharmacokinet Pharmacodyn 2022; 49:337-362. [DOI: 10.1007/s10928-021-09800-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 12/22/2021] [Indexed: 12/19/2022]
|
20
|
Peletier LA. An Extended Model Including Target Turnover, Ligand-Target Complex Kinetics, and Binding Properties to Describe Drug-Receptor Interactions. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2385:19-46. [PMID: 34888714 DOI: 10.1007/978-1-0716-1767-0_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Since the beginning of this century, target-mediated drug disposition has become a central concept in modeling drug action in drug development. It combines a range of processes, such as turnover, protein binding, internalization, and non-specific elimination, and often serves as a nucleus of more complex pharmacokinetic models. It is simple enough to comprehend but complex enough to be able to describe a wide range of phenomena and data sets. However, the complexity comes at a price: many parameters. In this chapter, we present an overview of the temporal development of the compounds involved after different types of drug doses and offer convenient handles for dissecting data sets in a sophisticated manner in order to estimate the values of these parameters, such as rate constants and pertinent concentrations.
Collapse
|
21
|
Wang J, Giragossian C, Hansel S. Analyze impact of tumor-associated kinetics on antibody delivery in solid tumors with a physiologically based pharmacokinetics/pharmacodynamics model. Eur J Pharm Biopharm 2021; 168:110-121. [PMID: 34478854 DOI: 10.1016/j.ejpb.2021.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/23/2021] [Accepted: 08/27/2021] [Indexed: 11/17/2022]
Abstract
Monoclonal antibody (mAb)-based drugs are critical anti-cancer therapies. Unfortunately, therapeutic efficacy can be compromised by spatially heterogeneous intratumoral Ab deposition. Binding-site barriers arising from Ab and tumor-associated kinetics often underlie this phenomenon. Quantitative insight into these issues may lead to more efficient drug delivery. Difficulties in addressing this issue include (1) lack of techniques to quantify critical kinetic events, (2) lack of a pharmacokinetic/pharmacodynamic (PK/PD) model to assess important parameters for specific tumor types, and (3) uncertainty or variability of critical kinetic factors even within a single tumor type. This study developed a mechanism-based PK/PD model to profile heterogeneous distribution of Ab within tumors and tested this model using real-life experimental data. Model simulations incorporating several uncertainties were used to determine how mAb and tumor-associated kinetics influence receptor occupancy. Simulations were also used to predict the potential impact of these findings in preclinical tumor models and human tumors. We found significant differences in tumor-associated kinetics between groups in which mAb therapy was effective versus groups in which it was ineffective. These kinetic differences included rates of tumor-associated antigen (TAA) degradation, TAA expression, apparent flow rates of interstitial fluid, and ratios of Ab-TAA complex internalization to TAA degradation. We found less significant differences in mAb kinetics, including rates of clearance or affinity for target antigens. In conclusion, our mechanism-based PK/PD model suggests that TAA-associated kinetic factors participate more significantly than those associated with the Ab in generating barriers to mAb delivery and distribution in tumors.
Collapse
Affiliation(s)
- Jun Wang
- Biotherapeutics Discovery Research, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA.
| | - Craig Giragossian
- Biotherapeutics Discovery Research, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| | - Steven Hansel
- Biotherapeutics Discovery Research, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| |
Collapse
|
22
|
Singh R, Moreno M, Stanimirovic D. Comparison of Various Approaches to Translate Non-Linear Pharmacokinetics of Monoclonal Antibodies from Cynomolgus Monkey to Human. Eur J Drug Metab Pharmacokinet 2021; 46:555-567. [PMID: 34120326 DOI: 10.1007/s13318-021-00691-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND AND OBJECTIVES The prediction of pharmacokinetics of monoclonal antibodies (mAbs) exhibiting non-linear pharmacokinetics in preclinical species to human is challenging, and very limited scientific work has been published in this field of research. Therefore, we have conducted an elaborate comparative assessment to determine the most reliable preclinical to clinical scaling strategy for mAbs with non-linear pharmacokinetics. METHODS We have compared three different scaling approaches to predict human pharmacokinetics from cynomolgus monkey. In the first approach, cynomolgus monkey pharmacokinetic parameters estimated using a two-compartment model with parallel linear and non-linear elimination were allometrically scaled to simulate human pharmacokinetics. In the second approach, allometric exponents were integrated with a minimal physiologically based pharmacokinetic (mPBPK) model to translate human pharmacokinetics. In the third approach, we have employed a species time-invariant method, wherein a two-compartment model with parallel linear and non-linear elimination was used as a framework model for simulation of the human profile. RESULTS Human exposure parameters projected by an integrated allometric method were only within two fold for approximately 45-70% of predictions at different doses of five mAbs evaluated, while approximately 70-80% of Cmax and AUC predictions by integrated mPBPK modelling as well as the species time-invariant method were within two-fold error. The average fold error for clearance predictions by the integrated mPBPK method was 1.10-1.45 fold, whilst for the species time-variant and integrated allometric methods, the average fold error was between 1.04 and 1.37 fold and 1.24 and 2.13 fold, respectively. CONCLUSIONS Our findings suggest that the species time-variant method and mPBPK proposed by us can be employed to reliably translate non-linear pharmacokinetics of mAbs from cynomolgus monkey to human.
Collapse
Affiliation(s)
- Renu Singh
- Translation Bioscience, Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada. .,Global DMPK, Research IVIVT, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA. .,Translation Bioscience, Human Health Therapeutics, National Research Council of Canada, Montreal, QC, Canada.
| | - Maria Moreno
- Translation Bioscience, Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada
| | - Danica Stanimirovic
- Translation Bioscience, Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada
| |
Collapse
|
23
|
Conner KP, Pastuskovas CV, Soto M, Thomas VA, Wagner M, Rock DA. Preclinical characterization of the ADME properties of a surrogate anti-IL-36R monoclonal antibody antagonist in mouse serum and tissues. MAbs 2021; 12:1746520. [PMID: 32310023 PMCID: PMC7188401 DOI: 10.1080/19420862.2020.1746520] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The decision to pursue a monoclonal antibody (mAb) as a therapeutic for disease intervention requires the assessment of many factors, such as target-biology, including the total target burden and its accessibility at the intended site of action, as well as mAb-specific properties like binding affinity and the pharmacokinetics in serum and tissue. Interleukin-36 receptor (IL-36 R) is a member of the IL-1 family cytokine receptors and an attractive target to treat numerous epithelial-mediated inflammatory conditions, including psoriatic and rheumatoid arthritis, asthma, and chronic obstructive pulmonary disease. However, information concerning the expression profile of IL-36 R at the protein level is minimal, so the feasibility of developing a therapeutic mAb against this target is uncertain. Here, we present a characterization of the properties associated with absorption, distribution, metabolism, and excretion of a high-affinity IL-36 R-targeted surrogate rat (IgG2a) mAb antagonist in preclinical mouse models. The presence of IL-36 R in the periphery was confirmed unequivocally as the driver of non-linear pharmacokinetics in blood/serum, although a predominant site of tissue accumulation was not observed based upon the kinetics of radiotracer. Additionally, the contribution of IL-36 R-mediated catabolism of mAb in kidney was tested in a 5/6 nephrectomized mouse model where minimal effects on serum pharmacokinetics were observed, although analysis of functional mAb in urine suggests that target can influence the amount of mAb excreted. Our data highlight an interesting case of target-mediated drug disposition (TMDD) where low, yet broadly expressed levels of membrane-bound target result in a cumulative effect to drive TMDD behavior typical of a large, saturable target sink. The potential differences between our mouse model and IL-36 R target profile in humans are also presented.
Collapse
Affiliation(s)
- Kip P Conner
- Department Pharmacokinetics and Drug Metabolism, Amgen, South San Francisco, CA, USA
| | - Cinthia V Pastuskovas
- Department Pharmacokinetics and Drug Metabolism, Amgen, South San Francisco, CA, USA
| | - Marcus Soto
- Department Pharmacokinetics and Drug Metabolism, Amgen, Thousand Oaks, CA, USA
| | - Veena A Thomas
- Department Pharmacokinetics and Drug Metabolism, Amgen, South San Francisco, CA, USA
| | - Mylo Wagner
- Department Pharmacokinetics and Drug Metabolism, Amgen, Thousand Oaks, CA, USA
| | - Dan A Rock
- Department Pharmacokinetics and Drug Metabolism, Amgen, South San Francisco, CA, USA
| |
Collapse
|
24
|
Xin Y, Xu F, Gao Y, Bhatt N, Chamberlain J, Sile S, Hammel S, Holt RJ, Ramanathan S. Pharmacokinetics and Exposure-Response Relationship of Teprotumumab, an Insulin-Like Growth Factor-1 Receptor-Blocking Antibody, in Thyroid Eye Disease. Clin Pharmacokinet 2021; 60:1029-1040. [PMID: 33768488 PMCID: PMC8332554 DOI: 10.1007/s40262-021-01003-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2021] [Indexed: 11/28/2022]
Abstract
Background and Objective Thyroid eye disease (TED) is characterized by inflammation/expansion of orbital tissues, proptosis, and diplopia. Teprotumumab is the first US Food and Drug Administration-approved therapy for TED, administered as an initial intravenous infusion of 10 mg/kg followed by 20 mg/kg every 3 weeks for an additional seven infusions. The objective of this article is to discuss the pharmacokinetics and exposure-response profile for teprotumumab in patients with TED. Methods A population pharmacokinetic analysis was performed to characterize pharmacokinetics and select dosing in patients with TED. Exposure-response was evaluated for efficacy (proptosis response, clinical activity score categorical response, and diplopia response) and safety (hyperglycemia, muscle spasms, and hearing impairment) parameters. Results Teprotumumab pharmacokinetics was linear in patients with TED, with low systemic clearance (0.334 L/day), low volume of distribution (3.9 and 4.2 L for the central and peripheral compartment, respectively), and a long elimination half-life (19.9 days). The approved dosing regimen provided > 20 µg/mL for > 90% insulin-like growth factor 1 receptor saturation throughout the dosing interval. Model-predicted mean (± standard deviation) steady-state area under the concentration-time curve, peak, and trough concentrations in patients with TED were 131 (± 30.9) mg∙h/mL, 643 (± 130) µg/mL, and 157 (± 50.6) µg/mL, respectively. Female patients had a 15% higher steady-state peak concentration but a similar steady-state area under the concentration-time curve vs male patients. No other covariates affected teprotumumab pharmacokinetics. No meaningful correlations between teprotumumab exposures and efficacy or safety parameters were observed. Conclusions Teprotumumab pharmacokinetics was well characterized in patients with TED, and generally consistent with other IgG1 antibodies. Efficacy was consistent across the exposure range with a well-tolerated safety profile supporting the current dose regimen for patients with TED. Supplementary Information The online version contains supplementary material available at 10.1007/s40262-021-01003-3.
Collapse
Affiliation(s)
- Yan Xin
- Horizon Therapeutics plc, 150 S. Saunders Rd, Lake Forest, IL, 60045, USA.
| | - Fengyan Xu
- Shanghai Qiangshi Information Technology Co., Ltd, Shanghai, China
| | - Yuying Gao
- Shanghai Qiangshi Information Technology Co., Ltd, Shanghai, China
| | - Nivedita Bhatt
- Horizon Therapeutics plc, 150 S. Saunders Rd, Lake Forest, IL, 60045, USA
| | - Jason Chamberlain
- Horizon Therapeutics plc, 150 S. Saunders Rd, Lake Forest, IL, 60045, USA
| | - Saba Sile
- Horizon Therapeutics plc, 150 S. Saunders Rd, Lake Forest, IL, 60045, USA
| | - Suzy Hammel
- Horizon Therapeutics plc, 150 S. Saunders Rd, Lake Forest, IL, 60045, USA
| | - Robert J Holt
- Horizon Therapeutics plc, 150 S. Saunders Rd, Lake Forest, IL, 60045, USA
| | - Srini Ramanathan
- Horizon Therapeutics plc, 150 S. Saunders Rd, Lake Forest, IL, 60045, USA
| |
Collapse
|
25
|
Tang X, Zeng X, Guan X, Chen R, Hu P. Modeling and Simulation to Support Phase Ib/IIa Dose Selection for WBP216, A Long Half-Life Fully Human Monoclonal Antibody Against Interleukin-6. Front Pharmacol 2021; 12:617265. [PMID: 33679400 PMCID: PMC7930490 DOI: 10.3389/fphar.2021.617265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/14/2021] [Indexed: 11/16/2022] Open
Abstract
WBP216 is an innovative IL-6 antibody, presenting high affinity to IL-6 and a long half-life (40–60 days). To optimize the dosage regimen for future clinical trials, pharmacokinetics (PK) and pharmacodynamics (PD) of WBP216 would be firstly characterized in Chinese rheumatoid arthritis (RA) patients. PK, CRP and DAS28 data of WBP216 were collected from 26 RA patients in a single ascending dose study. Non-linear mixed effects modeling was used for a population PK/PD analysis. A two-compartment model with a sequential zero-first order absorption and a first order elimination best described PK behavior of WBP216. Apparent systemic clearance was 0.015 L/h, central volume was 8.04 L. CRP as the fast-decreasing endpoint and DAS28 as the slow-reacting endpoint were both fitted well through an indirect response model. The baseline of ALT and free IL-6 were found associated with PK/PD parameters during covariates exploration. Simulation results confirmed that a loading dose regimen either of administration at weeks 0, 2, and 6 or doubling the maintenance dose level, followed by maintenance dosing of 75–150 mg every 8 weeks, was expected to provide a best risk/benefit ratio in future clinical studies. We hope this first PK/PD study of WBP216 in Chinese RA patients will help in the clinical development of WBP216 in future and provide a reference to the dosage optimization of similar antibodies with long half-life. Clinical Trial Registration:CTR20170306
Collapse
Affiliation(s)
- Xiange Tang
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Clinical PK and PD Investigation for Innovative Drugs, Beijing, China
| | - Xiaofeng Zeng
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiaoduo Guan
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Clinical PK and PD Investigation for Innovative Drugs, Beijing, China
| | - Rui Chen
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Clinical PK and PD Investigation for Innovative Drugs, Beijing, China
| | - Pei Hu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Clinical PK and PD Investigation for Innovative Drugs, Beijing, China
| |
Collapse
|
26
|
Chung TK, Lee HA, Park SI, Oh DY, Lee KW, Kim JW, Kim JH, Woo A, Lee SJ, Bang YJ, Lee H. A target-mediated drug disposition population pharmacokinetic model of GC1118, a novel anti-EGFR antibody, in patients with solid tumors. Clin Transl Sci 2021; 14:990-1001. [PMID: 33382918 PMCID: PMC8212746 DOI: 10.1111/cts.12963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/18/2020] [Accepted: 12/03/2020] [Indexed: 11/29/2022] Open
Abstract
Abstract GC1118 is a monoclonal antibody for epidermal growth factor receptor (EGFR) that is currently under clinical development to treat patients with solid tumors. In this study, the pharmacokinetics (PKs) of GC1118 were modeled in solid tumor patients who received a 2‐h intravenous infusion of GC1118 at 0.3, 1, 3, 5, or 4 mg/kg once‐weekly (Q1W) on days 1, 8, 15, and 22 or 8 mg/kg every other week on days 1 and 15. A target‐mediated drug disposition population PK model adequately described the concentration‐time profiles of GC1118. Monte‐Carlo simulation experiments of the PK profiles and EGFR occupancies (ROs) by GC1118 based on the final model showed that Q1W at 4 or 5 mg/kg will produce a better antitumor effect than Q2W at 8 mg/kg. Because GC1118 was safer at 4 mg/kg than 5 mg/kg in the phase I study, we suggest to test the 4 mg/kg Q1W regimen in further clinical trials with GC1118. Study Highlights WHAT IS THE CURRENT KNOWLEDGE ON THE TOPIC?
GC1118, a fully human IgG1 monoclonal antibody (mAb) for epidermal growth factor receptor (EGFR), showed a nonlinear pharmacokinetic (PK) profile in monkeys and humans. The total clearance of GC1118 decreased as the dose was increased up to 3–4 mg/kg in humans, beyond which it remained stable. The recommended phase II dose for GC1118 was 4 mg/kg intravenously infused over 2 h once weekly.
WHAT QUESTION DID THIS STUDY ADDRESS?
We developed a target‐mediated drug disposition (TMDD) population PK model that described the nonlinear PK profile of GC1118 in patients with solid tumors. We also simulated the PK profiles and receptor occupancies for different dosage regimens.
WHAT DOES THIS STUDY ADD TO OUR KNOWLEDGE?
The TMDD population PK model adequately described the nonlinear and multiphasic PK profiles of GC1118 in humans. The simulation experiment showed that once‐weekly GC1118 at 4–5 mg/kg could be more efficacious than the biweekly regimen at 8 mg/kg.
HOW MIGHT THIS CHANGE CLINICAL PHARMACOLOGY OR TRANSLATIONAL SCIENCE?
The pharmacometrics analysis could support better informed drug development decisions for GC1118, particularly for determining an optimal dosage regimen.
Collapse
Affiliation(s)
- Tae Kyu Chung
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Hyun A Lee
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Sang-In Park
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Korea
| | - Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Keun-Wook Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jin Won Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | | | | | - Yung-Jue Bang
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Howard Lee
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea.,Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea.,Center for Convergence Approaches in Drug Development, Seoul National University, Seoul, Korea.,Advanced Institute of Convergence Technology, Suwon, Korea
| |
Collapse
|
27
|
Hijazi Y. Prediction of Half-Life Extension of Peptides via Serum Albumin Binding: Current Challenges. Eur J Drug Metab Pharmacokinet 2021; 46:163-172. [PMID: 33386550 DOI: 10.1007/s13318-020-00664-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2020] [Indexed: 10/22/2022]
Abstract
The development of peptide therapeutics has increased enormously in recent decades. Many of the peptide drugs and antibody fragments that lack Fc backbone have a short half-life in circulation. In general, the half-life supports the design of the dosing regimen and frequency of administration, which are key aspects in the discovery of peptide drugs intended for long duration of action. Less frequent administration such as weekly or monthly can improve compliance and adherence to therapy. Serum albumin binding is a key approach to extend the half-life of peptide drugs. Despite the evidence of half-life prolongation of a variety of peptide drugs via albumin, quantitative prediction for humans is still a key question. Challenges in the measurement of albumin binding and in understanding the clearance mechanisms can limit quantitative prediction. We integrated pharmacokinetic concepts and albumin binding across species in a quantitative model to be used as a tool for prediction of half-life. Preliminary validation on a limited dataset indicated a good correlation between predicted and observed values. Further development of more quantitative models is warranted.
Collapse
Affiliation(s)
- Youssef Hijazi
- R&D, Drug Metabolism and Pharmacokinetics, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H 831, C 0442, 65926, Frankfurt, Germany.
| |
Collapse
|
28
|
Ponte JF, Lanieri L, Khera E, Laleau R, Ab O, Espelin C, Kohli N, Matin B, Setiady Y, Miller ML, Keating TA, Chari R, Pinkas J, Gregory R, Thurber GM. Antibody Co-Administration Can Improve Systemic and Local Distribution of Antibody-Drug Conjugates to Increase In Vivo Efficacy. Mol Cancer Ther 2021; 20:203-212. [PMID: 33177153 PMCID: PMC7790875 DOI: 10.1158/1535-7163.mct-20-0451] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/06/2020] [Accepted: 10/27/2020] [Indexed: 11/16/2022]
Abstract
Several antibody-drug conjugates (ADC) showing strong clinical responses in solid tumors target high expression antigens (HER2, TROP2, Nectin-4, and folate receptor alpha/FRα). Highly expressed tumor antigens often have significant low-level expression in normal tissues, resulting in the potential for target-mediated drug disposition (TMDD) and increased clearance. However, ADCs often do not cross-react with normal tissue in animal models used to test efficacy (typically mice), and the impact of ADC binding to normal tissue antigens on tumor response remains unclear. An antibody that cross-reacts with human and murine FRα was generated and tested in an animal model where the antibody/ADC bind both human tumor FRα and mouse FRα in normal tissue. Previous work has demonstrated that a "carrier" dose of unconjugated antibody can improve the tumor penetration of ADCs with high expression target-antigens. A carrier dose was employed to study the impact on cross-reactive ADC clearance, distribution, and efficacy. Co-administration of unconjugated anti-FRα antibody with the ADC-improved efficacy, even in low expression models where co-administration normally lowers efficacy. By reducing target-antigen-mediated clearance in normal tissue, the co-administered antibody increased systemic exposure, improved tumor tissue penetration, reduced target-antigen-mediated uptake in normal tissue, and increased ADC efficacy. However, payload potency and tumor antigen saturation are also critical to efficacy, as shown with reduced efficacy using too high of a carrier dose. The judicious use of higher antibody doses, either through lower DAR or carrier doses, can improve the therapeutic window by increasing efficacy while lowering target-mediated toxicity in normal tissue.
Collapse
Affiliation(s)
| | | | - Eshita Khera
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| | | | - Olga Ab
- ImmunoGen, Waltham, Massachusetts
| | | | | | | | | | | | | | | | | | | | - Greg M Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
29
|
Xiang H, Liu L, Gao Y, Ahene A, Macal M, Hsu AW, Dreiling L, Collins H. Population pharmacokinetic analysis of phase 1 bemarituzumab data to support phase 2 gastroesophageal adenocarcinoma FIGHT trial. Cancer Chemother Pharmacol 2020; 86:595-606. [PMID: 32965540 PMCID: PMC7561547 DOI: 10.1007/s00280-020-04139-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 09/06/2020] [Indexed: 12/11/2022]
Abstract
Purpose To report population pharmacokinetic (PK) analysis of the phase 1 study (FPA144-001, NCT02318329) and to select a clinical dose and schedule that will achieve an empirical target trough concentration (Ctrough) for an anti-fibroblast growth factor receptor 2b antibody, bemarituzumab. Methods Nonlinear mixed-effect modeling was used to analyse PK data. In vitro binding affinity and receptor occupancy of bemarituzumab were determined. Simulation was conducted to estimate dose and schedule to achieve an empirical target Ctrough in a phase 2 trial (FIGHT, NCT03694522) for patients receiving first-line treatment combined with modified 5-fluourouracil, oxaliplatin and leucovorin (mFOLFOX6) for gastric and gastroesophageal junction adenocarcinoma. Results Bemarituzumab PK is best described by a two-compartment model with parallel linear and nonlinear (Michaelis–Menten) elimination from the central compartment. Albumin, gender, and body weight were identified as the covariates on the linear clearance and/or volume of distribution in the central compartment, and no dose adjustment was warranted. An empirical target of bemarituzumab Ctrough of ≥ 60 µg/mL was projected to achieve > 95% receptor occupancy based on in vitro data. Fifteen mg/kg every 2 weeks, with a single dose of 7.5 mg/kg on Cycle 1 Day 8, was projected to achieve the target Ctrough on Day 15 in 98% of patients with 96% maintaining the target at steady state, which was confirmed in the FIGHT trial. Conclusion A projected dose and schedule to achieve the target Ctrough was validated in phase 1 of the FIGHT trial which supported selection of the phase 2 dose and schedule for bemarituzumab. Electronic supplementary material The online version of this article (10.1007/s00280-020-04139-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hong Xiang
- Five Prime Therapeutics, Inc., 111 Oyster Point Blvd, South San Francisco, CA, 94080, USA.
| | - Lucy Liu
- Shanghai Qiangshi Information Technology Co., Ltd, Shanghai, China
| | - Yuying Gao
- Shanghai Qiangshi Information Technology Co., Ltd, Shanghai, China
| | - Ago Ahene
- Five Prime Therapeutics, Inc., 111 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Monica Macal
- Five Prime Therapeutics, Inc., 111 Oyster Point Blvd, South San Francisco, CA, 94080, USA.,TRex Bio, Inc., South San Francisco, CA, USA
| | - Amy W Hsu
- Five Prime Therapeutics, Inc., 111 Oyster Point Blvd, South San Francisco, CA, 94080, USA.,Merck and Co., South San Francisco, CA, USA
| | - Lyndah Dreiling
- Five Prime Therapeutics, Inc., 111 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Helen Collins
- Five Prime Therapeutics, Inc., 111 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| |
Collapse
|
30
|
Longuespée R, Theile D, Fresnais M, Burhenne J, Weiss J, Haefeli WE. Approaching sites of action of drugs in clinical pharmacology: New analytical options and their challenges. Br J Clin Pharmacol 2020; 87:858-874. [PMID: 32881012 DOI: 10.1111/bcp.14543] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/20/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022] Open
Abstract
Clinical pharmacology is an important discipline for drug development aiming to define pharmacokinetics (PK), pharmacodynamics (PD) and optimum exposure to drugs, i.e. the concentration-response relationship and its modulators. For this purpose, information on drug concentrations at the anatomical, cellular and molecular sites of action is particularly valuable. In pharmacological assays, the limited accessibility of target cells in readily available samples (i.e. blood) often hampers mass spectrometry-based monitoring of the absolute quantity of a compound and the determination of its molecular action at the cellular level. Recently, new sample collection methods have been developed for the specific capture of rare circulating cells, especially for the diagnosis of circulating tumour cells. In parallel, new advances and developments in mass spectrometric instrumentation now allow analyses to be scaled down to the cellular level. Together, these developments may permit the monitoring of minute drug quantities and show their effect at the cellular level. In turn, such PK/PD associations on a cellular level would not only enrich our pharmacological knowledge of a given compound but also expand the basis for PK/PD simulations. In this review, we describe novel concepts supporting clinical pharmacology at the anatomical, cellular and molecular sites of action, and highlight the new challenges in mass spectrometry-based monitoring. Moreover, we present methods to tackle these challenges and define future needs.
Collapse
Affiliation(s)
- Rémi Longuespée
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Dirk Theile
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Margaux Fresnais
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital of Heidelberg, Heidelberg, Germany.,German Cancer Consortium (DKTK)-German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen Burhenne
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Johanna Weiss
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Walter E Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital of Heidelberg, Heidelberg, Germany
| |
Collapse
|
31
|
Hangasky JA, Waldmann TA, Santi DV. Interleukin 15 Pharmacokinetics and Consumption by a Dynamic Cytokine Sink. Front Immunol 2020; 11:1813. [PMID: 32903632 PMCID: PMC7438588 DOI: 10.3389/fimmu.2020.01813] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Interleukin-15 (IL-15) is crucial for the proliferation and survival of NK and CD8+ T memory cells, and of significant interest in immuno-oncology. Immune cell expansion requires continuous IL-15 exposure above a threshold concentration for an extended period. However, the short t1/2 of IL-15 makes this impossible to achieve after a single injection without a high Cmax and toxicities. The most effective way to deliver IL-15 is continuous intra-venous infusion, but this administration mode is impractical. Efforts have been devoted to developing IL-15 agonists which after a single injection maintain the cytokine in a narrow therapeutic window for a long period. Enigmatically, although the half-life extension technologies used often extend the half-life of a protein to 1 or more weeks, the modified IL-15 agonists studied usually have systemic elimination half-lives of only a few hours and rarely much longer than 1 day. These short half-lives—common to all circulating IL-15 agonists thus far reported—can be explained by a dynamic increase in clearance of the agonists that accompanies target immune cell proliferation. What is needed is an IL-15 agonist that is as effective as continuous intravenous infusion, but with the convenience and acceptance of single injections at 1-week or longer intervals.
Collapse
Affiliation(s)
| | - Thomas A Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, United States
| | | |
Collapse
|
32
|
Smart K, Bröske A, Rüttinger D, Mueller C, Phipps A, Walz A, Ries C, Baehner M, Cannarile M, Meneses‐Lorente G. PK/PD Mediated Dose Optimization of Emactuzumab, a CSF1R Inhibitor, in Patients With Advanced Solid Tumors and Diffuse-Type Tenosynovial Giant Cell Tumor. Clin Pharmacol Ther 2020; 108:616-624. [PMID: 32575160 PMCID: PMC7589268 DOI: 10.1002/cpt.1964] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/04/2020] [Indexed: 01/03/2023]
Abstract
Targeted biological therapies may achieve maximal therapeutic efficacy at doses below the maximum tolerated dose (MTD); therefore, the search for the MTD in clinical studies may not be ideal for these agents. Emactuzumab is an investigational monoclonal antibody that binds to and inhibits the activation of the cell surface colony‐stimulating factor‐1 receptor. Here, we show how modeling target‐mediated drug disposition coupled with pharmacodynamic end points was used to optimize the dose of emactuzumab without defining an MTD. The model could be used to recommend doses across different disease indications. The approach recommended an optimal biological dose of emactuzumab for dosing every 2 weeks (q2w) ≥ 900 mg, approximately three‐fold lower than the highest dose tested clinically. The model predicted that emactuzumab doses ≥ 900 mg q2w would achieve target saturation in excess of 90% over the entire dosing cycle. Subsequently, a dose of 1,000 mg q2w was used in the extension phase of a phase I study of emactuzumab in patients with advanced solid tumors or diffuse‐type tenosynovial giant cell tumor. Clinical data from this study were consistent with model predictions. The model was also used to predict the optimum dose of emactuzumab for use with dosing every 3 weeks, enabling dosing flexibility with respect to comedications. In summary, this work demonstrates the value of quantitative clinical pharmacology approaches to dose selection in oncology as opposed to traditional MTD methods.
Collapse
MESH Headings
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/pharmacokinetics
- Clinical Trials, Phase I as Topic
- Drug Administration Schedule
- Drug Dosage Calculations
- Giant Cell Tumor of Tendon Sheath/drug therapy
- Giant Cell Tumor of Tendon Sheath/metabolism
- Giant Cell Tumor of Tendon Sheath/pathology
- Humans
- Models, Biological
- Molecular Targeted Therapy
- Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors
- Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
- Signal Transduction
- Treatment Outcome
Collapse
Affiliation(s)
- Kevin Smart
- Roche Innovation Center WelwynWelwyn Garden CityUK
| | | | | | | | - Alex Phipps
- Roche Innovation Center WelwynWelwyn Garden CityUK
| | | | - Carola Ries
- Roche Innovation Center MunichPenzbergGermany
| | | | | | | |
Collapse
|
33
|
Wang X, Du W, Zhang X, Li P. The Influence of Different Disease States on Rituximab Pharmacokinetics. Curr Drug Metab 2020; 21:938-946. [PMID: 32682367 DOI: 10.2174/1389200221666200719004035] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/04/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND The anti-CD20 antibody rituximab, which promotes the selective depletion of CD20 positive B cells, was the first targeted therapy that was approved for the treatment of B-cell malignancies, and it is now widely prescribed in both malignant and non-malignant, immune-related diseases. However, the cause of its various clinical responses in certain diseases, have not been clearly elucidated. The variabilities in inter-individual pharmacokinetic and the emerging evidence of the relationships between pharmacokinetic and pharmacodynamic may provide a better understanding of this drug. METHODS We searched and summarized the latest published articles on rituximab pharmacokinetic profiles and the pharmacokinetic/pharmacodynamic models in different patient populations, including B-cell malignancies, rheumatoid arthritis, ANCA-associated vasculitis, and glomerular kidney diseases. RESULTS Most pharmacokinetic data are drawn from clinical studies in oncology clinical practice. Body weight, gender, and antigen-related factors are proven to be the key factors affecting rituximab pharmacokinetics. In addition, the positive exposure-response relations were reported, which provide encouraging evidence for individualized therapies. While in immune disorders, especially in the off-labeled indications, pharmacokinetic studies are quite limited. Compared with that in B-cell malignancies, the differences in the pharmacokinetic parameters may be attributed to the different pathogeneses of diseases, mechanisms of action and dosing strategies. However, the correlation between drug exposure and clinical outcomes remains unclear. CONCLUSION Here, we provide an overview of the complexities associated with rituximab pharmacokinetics and pharmacodynamics in different diseases. Although many influencing factors need to be verified in future studies, a better understanding of the relationships between pharmacokinetic and pharmacodynamic may assist in optimizing rituximab clinical practice.
Collapse
Affiliation(s)
- Xiaoxing Wang
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| | - Wenwen Du
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xianglin Zhang
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| | - Pengmei Li
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
34
|
Glassman PM, Muzykantov VR. Target-mediated exposure enhancement: a previously unexplored limit of TMDD. J Pharmacokinet Pharmacodyn 2020; 47:411-420. [PMID: 32488574 DOI: 10.1007/s10928-020-09693-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 05/28/2020] [Indexed: 11/25/2022]
Abstract
Target-mediated drug disposition (TMDD) is often observed for targeted therapeutics, and manifests as decreases in clearance and volume of distribution with increasing dose as a result of saturable, high affinity target binding. In the present work, we demonstrate that classically defined TMDD is just one of the characteristic features of the system. In fact, for molecules with rapid non-specific elimination relative to target-mediated elimination, binding to target may actually lead to improved exposure at sub-saturating doses. This feature, which we refer to as target-mediated exposure enhancement (TMEE), produces the opposite trend to classical TMDD, i.e., with increasing dose levels, clearance and volume of distribution will also increase. The general model of TMDD was able to well-characterize the pharmacokinetics of two molecules that display TMEE, ALX-0081 and linagliptin. Additional fittings using the commonly reported TMDD model approximations revealed that both the quasi-equilibrium and quasi-steady-state approximations were able to well-describe TMEE; however, the Michaelis-Menten approximation was unable to describe this behavior. With the development of next-generation therapeutics with high affinity for target and rapid non-specific elimination, such as antibody fragments and peptides, this previously unexplored limit of TMDD is anticipated to become increasingly relevant for describing pharmacokinetics of investigational therapeutics.
Collapse
Affiliation(s)
- Patrick M Glassman
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Bldg 421, 10-178 Translational Research Center, Philadelphia, PA, 19104-5158, USA.
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Bldg 421, 10-178 Translational Research Center, Philadelphia, PA, 19104-5158, USA
| |
Collapse
|
35
|
A clinical population pharmacokinetic/pharmacodynamic model for BIIB059, a monoclonal antibody for the treatment of systemic and cutaneous lupus erythematosus. J Pharmacokinet Pharmacodyn 2020; 47:255-266. [PMID: 32335844 DOI: 10.1007/s10928-020-09688-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 04/15/2020] [Indexed: 10/24/2022]
Abstract
A population pharmacokinetic/pharmacodynamic (popPK/PD) model for BIIB059 (anti-blood dendritic cell antigen 2 [anti-BDCA2]), a humanized immunoglobulin G1 monoclonal antibody currently under development for the treatment of SLE and CLE, is presented. BIIB059 binds BDCA2, a plasmacytoid dendritic cell (pDC)-specific receptor that inhibits the production of IFN-I and other inflammatory mediators when ligated. Phase 1 PK and PD data of healthy adult volunteers (HV, n = 87) and SLE subjects (n = 22) were utilized for the development of the popPK/PD model. The data included single and multiple dosing of intravenous and subcutaneous BIIB059. BDCA2 internalization (PD marker) was measured for all subjects by monitoring reduction of BDCA2 on pDC cell surface and used for development of the popPD model. A two-compartment popPK model with linear plus non-linear elimination was found to best describe BIIB059 PK. BDCA2 levels were best captured using an indirect response model with stimulation of the elimination of BDCA2. Clearance in SLE subjects was 25% higher compared to HV (6.87 vs 5.52 mL/h). Bodyweight was identified as only other covariate on clearance and central volume. The estimates of EC50 and Emax were 0.35 μg/mL and 8.92, respectively. No difference in EC50 and Emax was observed between SLE and HV. The popPK/PD model described the data accurately, as evaluated by pcVPCs and bootstrap. The presented popPK/PD model for BIIB059 provides valuable insight into the dynamics and dose-response relationship of BIIB059 for the treatment of SLE and CLE and was used to guide dose selection for the Phase 2 clinical study (NCT02847598).
Collapse
|
36
|
Clarelli F, Liang J, Martinecz A, Heiland I, Abel Zur Wiesch P. Multi-scale modeling of drug binding kinetics to predict drug efficacy. Cell Mol Life Sci 2020; 77:381-394. [PMID: 31768605 PMCID: PMC7010620 DOI: 10.1007/s00018-019-03376-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/06/2019] [Accepted: 11/12/2019] [Indexed: 01/18/2023]
Abstract
Optimizing drug therapies for any disease requires a solid understanding of pharmacokinetics (the drug concentration at a given time point in different body compartments) and pharmacodynamics (the effect a drug has at a given concentration). Mathematical models are frequently used to infer drug concentrations over time based on infrequent sampling and/or in inaccessible body compartments. Models are also used to translate drug action from in vitro to in vivo conditions or from animal models to human patients. Recently, mathematical models that incorporate drug-target binding and subsequent downstream responses have been shown to advance our understanding and increase predictive power of drug efficacy predictions. We here discuss current approaches of modeling drug binding kinetics that aim at improving model-based drug development in the future. This in turn might aid in reducing the large number of failed clinical trials.
Collapse
Affiliation(s)
- Fabrizio Clarelli
- Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, 9037, Tromsø, Norway
| | - Jingyi Liang
- Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, 9037, Tromsø, Norway
| | - Antal Martinecz
- Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, 9037, Tromsø, Norway
| | - Ines Heiland
- Department of Arctic and Marine Biology, UiT The Arctic University of Norway, 9037, Tromsø, Norway
| | - Pia Abel Zur Wiesch
- Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, 9037, Tromsø, Norway.
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, Blindern, P.O. Box 1137, 0318, Oslo, Norway.
| |
Collapse
|
37
|
Saruta M, Park DI, Kim YH, Yang SK, Jang BI, Cheon JH, Im JP, Kanai T, Katsuno T, Ishiguro Y, Nagaoka M, Isogawa N, Li Y, Banerjee A, Ahmad A, Hassan-Zahraee M, Clare R, Gorelick KJ, Cataldi F, Watanabe M, Hibi T. Anti-MAdCAM-1 antibody (PF-00547659) for active refractory Crohn's disease in Japanese and Korean patients: the OPERA study. Intest Res 2020; 18:45-55. [PMID: 32013314 PMCID: PMC7000638 DOI: 10.5217/ir.2019.00039] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 11/15/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND/AIMS PF-00547659 is a monoclonal antibody against human mucosal addressin cell adhesion molecule-1 (MAdCAM-1) that prevents the binding of α4β7+ lymphocytes to MAdCAM-expressing sites in the gastrointestinal tract with high affinity and selectivity, and is being developed for the treatment of Crohn's disease (CD). METHODS OPERA is a randomized, multicenter, double-blind, placebo-controlled study to investigate the efficacy, safety, and pharmacokinetics of PF-00547659 following subcutaneous administration in subjects with active CD, a history of failure or intolerance to anti-tumor necrosis factor and/or immunosuppressants, high-sensitivity C-reactive protein > 3.0 mg/L, and ulcers on colonoscopy. The primary endpoint was Crohn's Disease Activity Index-70 response at week 8 or 12. Subpopulation analyses for Asian subjects were performed as some differences are observed in genetics and clinical phenotypes in Asian CD patients compared with Western patients. RESULTS In this study, 265 CD subjects were randomized, with a subpopulation of 21 subjects (8 Japanese and 13 Korean) defined as the Asian population. In the overall and Asian populations; PF-00547659 was pharmacologically active as evidenced by soluble MAdCAM and circulating β7+ central memory CD4+ T-lymphocytes, although no clear evidence of efficacy was observed in any clinical endpoints; pharmacokinetics of PF-00547659 in the Asian subpopulation was generally comparable to the overall population; and the safety profile of PF-00547659 appeared acceptable up to 12 weeks of treatment. CONCLUSIONS In the overall and Asian populations, efficacy of PF-00547659 could not be demonstrated using any clinical endpoints compared with placebo. Pharmacokinetics and safety of PF-00547659 were generally comparable. Further studies with larger numbers of patients are required to confirm our results. (Trial Registration Number: NCT01276509).
Collapse
Affiliation(s)
- Masayuki Saruta
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Dong Il Park
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young-Ho Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Suk-Kyun Yang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Byung-Ik Jang
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Jae Hee Cheon
- Institute of Gastroenterology and Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Pil Im
- Division of Gastroenterology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Takanori Kanai
- Department of Internal Medicine, Keio University Hospital, Tokyo, Japan
| | - Tatsuro Katsuno
- Department of Gastroenterology, Chiba University Hospital, Chiba, Japan
| | - Yoh Ishiguro
- Department of Gastroenterology and Hematology, National Hospital Organization, Hirosaki National Hospital, Hirosaki, Japan
| | | | - Naoki Isogawa
- Clinical Statistics, Pfizer Japan Inc., Tokyo, Japan
| | - Yinhua Li
- Clinical Pharmacology, Development Japan, Pfizer Japan Inc., Tokyo, Japan
| | | | - Alaa Ahmad
- Gastroenterology, Pfizer Inc., Cambridge, MA, USA
| | | | - Robert Clare
- Biotherapeutics Clinical R&D, Pfizer Inc., Collegeville, PA, USA
| | | | - Fabio Cataldi
- Gastroenterology, Clinical Programs, Pfizer Inc., Cambridge, MA, USA
| | - Mamoru Watanabe
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshifumi Hibi
- Center for Advanced IBD Research and Treatment, Kitasato Institute Hospital, Kitasato University, Tokyo, Japan
| |
Collapse
|
38
|
Singh AP, Guo L, Verma A, Wong GGL, Thurber GM, Shah DK. Antibody Coadministration as a Strategy to Overcome Binding-Site Barrier for ADCs: a Quantitative Investigation. AAPS JOURNAL 2020; 22:28. [PMID: 31938899 DOI: 10.1208/s12248-019-0387-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/04/2019] [Indexed: 12/14/2022]
Abstract
It has been proposed that the binding-site barrier (BSB) for antibody-drug conjugates (ADCs) can be overcome with the help of antibody coadministration. However, broad utility of this strategy remains in question. Consequently, here, we have conducted in vivo experiments and pharmacokinetics-pharmacodynamics (PK-PD) modeling and simulation (M&S) to further evaluate the antibody coadministration hypothesis in a quantitative manner. Two different Trastuzumab-based ADCs, T-DM1 (no bystander effect) and T-vc-MMAE (with a bystander effect), were evaluated in high-HER2 (N87) and low-HER2 (MDA-MB-453) expressing tumors, with or without the coadministration of 1, 3, or 8-fold higher Trastuzumab. The tumor growth inhibition (TGI) data was quantitatively characterized using a semi-mechanistic PK-PD model to determine the nature of drug interaction for each coadministration regimen, by estimating the interaction parameter ψ. It was found that the coadministration strategy improved ADC efficacy under certain conditions and had no impact on ADC efficacy in others. The benefit was more pronounced for N87 tumors with very high antigen expression levels where the effect on treatment was synergistic (a synergistic drug interaction, ψ = 2.86 [2.6-3.12]). The benefit was diminished in tumor with lower antigen expression (MDA-MB-453) and payload with bystander effect. Under these conditions, the coadministration regimens resulted in an additive or even less than additive benefit (ψ ≤ 1). As such, our results suggest that while antibody coadministration may be helpful for ADCs in certain circumstances, one should not broadly apply this strategy to all the scenarios without first identifying the costs and benefits of this approach.
Collapse
Affiliation(s)
- Aman P Singh
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Leiming Guo
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Ashwni Verma
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Gloria Gao-Li Wong
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, New York, 14214-8033, USA
| | - Greg M Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA.
| |
Collapse
|
39
|
Lorentz CU, Verbout NG, Wallisch M, Hagen MW, Shatzel JJ, Olson SR, Puy C, Hinds MT, McCarty OJT, Gailani D, Gruber A, Tucker EI. Contact Activation Inhibitor and Factor XI Antibody, AB023, Produces Safe, Dose-Dependent Anticoagulation in a Phase 1 First-In-Human Trial. Arterioscler Thromb Vasc Biol 2020; 39:799-809. [PMID: 30700130 DOI: 10.1161/atvbaha.118.312328] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Objective- Factor XI (FXI) contributes to thrombotic disease while playing a limited role in normal hemostasis. We generated a unique, humanized anti-FXI antibody, AB023, which blocks factor XIIa-mediated FXI activation without inhibiting FXI activation by thrombin or the procoagulant function of FXIa. We sought to confirm the antithrombotic activity of AB023 in a baboon thrombosis model and to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics in healthy adult subjects. Approach and Results- In a primate model of acute vascular graft thrombosis, AB023 reduced platelet and fibrin accumulation within the grafts by >75%. To evaluate the safety of AB023, we performed a first-in-human study in healthy adult volunteers without any serious adverse events. Overall, 10 of 21 (48%) subjects experienced 20 treatment-emergent adverse events, with 7 of 16 (44%) subjects following active treatment and 3 of 5 (60%) subjects following placebo. AB023 did not increase bleeding or prothrombin times. Anticoagulation was verified by a saturable ≈2-fold prolongation of the partial thromboplastin time for over 1 month after the highest dose. Conclusions- AB023, which inhibits contact activation-initiated blood coagulation in vitro and experimental thrombus formation in primates, produced a dose-dependent duration of limited anticoagulation without drug-related adverse effects in a phase 1 trial. When put in context with earlier observations suggesting that FXI contributes to venous thromboembolism and cardiovascular disease, although contributing minimally to hemostasis, our data further justify clinical evaluation of AB023 in conditions where contact-initiated FXI activation is suspected to have a pathogenic role. Clinical Trial Registration- URL: http://www.clinicaltrials.gov . Unique identifier: NCT03097341.
Collapse
Affiliation(s)
- Christina U Lorentz
- From Aronora, Inc, Portland, OR (C.U.L., N.G.V., M.W., A.G., E.I.T.).,Department of Biomedical Engineering (C.U.L., N.G.V., M.W., M.W.H., J.J.S., S.R.O., C.P., M.T.H., O.J.T.M., A.G., E.I.T.), Oregon Health & Science University, Portland
| | - Norah G Verbout
- From Aronora, Inc, Portland, OR (C.U.L., N.G.V., M.W., A.G., E.I.T.).,Department of Biomedical Engineering (C.U.L., N.G.V., M.W., M.W.H., J.J.S., S.R.O., C.P., M.T.H., O.J.T.M., A.G., E.I.T.), Oregon Health & Science University, Portland
| | - Michael Wallisch
- From Aronora, Inc, Portland, OR (C.U.L., N.G.V., M.W., A.G., E.I.T.).,Department of Biomedical Engineering (C.U.L., N.G.V., M.W., M.W.H., J.J.S., S.R.O., C.P., M.T.H., O.J.T.M., A.G., E.I.T.), Oregon Health & Science University, Portland
| | - Matthew W Hagen
- Department of Biomedical Engineering (C.U.L., N.G.V., M.W., M.W.H., J.J.S., S.R.O., C.P., M.T.H., O.J.T.M., A.G., E.I.T.), Oregon Health & Science University, Portland
| | - Joseph J Shatzel
- Department of Biomedical Engineering (C.U.L., N.G.V., M.W., M.W.H., J.J.S., S.R.O., C.P., M.T.H., O.J.T.M., A.G., E.I.T.), Oregon Health & Science University, Portland.,Division of Hematology and Medical Oncology (J.J.S., S.R.O., O.J.T.M., A.G.), Oregon Health & Science University, Portland
| | - Sven R Olson
- Department of Biomedical Engineering (C.U.L., N.G.V., M.W., M.W.H., J.J.S., S.R.O., C.P., M.T.H., O.J.T.M., A.G., E.I.T.), Oregon Health & Science University, Portland.,Division of Hematology and Medical Oncology (J.J.S., S.R.O., O.J.T.M., A.G.), Oregon Health & Science University, Portland
| | - Cristina Puy
- Department of Biomedical Engineering (C.U.L., N.G.V., M.W., M.W.H., J.J.S., S.R.O., C.P., M.T.H., O.J.T.M., A.G., E.I.T.), Oregon Health & Science University, Portland
| | - Monica T Hinds
- Department of Biomedical Engineering (C.U.L., N.G.V., M.W., M.W.H., J.J.S., S.R.O., C.P., M.T.H., O.J.T.M., A.G., E.I.T.), Oregon Health & Science University, Portland
| | - Owen J T McCarty
- Department of Biomedical Engineering (C.U.L., N.G.V., M.W., M.W.H., J.J.S., S.R.O., C.P., M.T.H., O.J.T.M., A.G., E.I.T.), Oregon Health & Science University, Portland.,Division of Hematology and Medical Oncology (J.J.S., S.R.O., O.J.T.M., A.G.), Oregon Health & Science University, Portland
| | - David Gailani
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN (D.G.)
| | - András Gruber
- From Aronora, Inc, Portland, OR (C.U.L., N.G.V., M.W., A.G., E.I.T.).,Department of Biomedical Engineering (C.U.L., N.G.V., M.W., M.W.H., J.J.S., S.R.O., C.P., M.T.H., O.J.T.M., A.G., E.I.T.), Oregon Health & Science University, Portland.,Division of Hematology and Medical Oncology (J.J.S., S.R.O., O.J.T.M., A.G.), Oregon Health & Science University, Portland
| | - Erik I Tucker
- From Aronora, Inc, Portland, OR (C.U.L., N.G.V., M.W., A.G., E.I.T.).,Department of Biomedical Engineering (C.U.L., N.G.V., M.W., M.W.H., J.J.S., S.R.O., C.P., M.T.H., O.J.T.M., A.G., E.I.T.), Oregon Health & Science University, Portland
| |
Collapse
|
40
|
Viala M, Tosi D. [Determining the dose to be injected in the first clinical trials with monoclonal antibodies: not so easy!]. Med Sci (Paris) 2020; 35:1121-1129. [PMID: 31903926 DOI: 10.1051/medsci/2019209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Monoclonal antibodies are a therapeutic tool frequently used in oncology, as they allow the specific targeting of molecules expressed by cancer cells and, in most cases, induce minimal toxic effects on healthy tissues. Because monoclonal antibodies frequently lack significant toxicity and are not associated to a direct relationship between dose and effect, the methods of clinical development traditionally used for chemotherapy agents are scarcely useful for this class of drugs. In addition, no consensus exists on the definition of parameters different from toxicity that could assist the process of dose selection of monoclonal antibody in early clinical trials.
Collapse
Affiliation(s)
- Marie Viala
- Unité d'Essais de Phase Précoce (UEPP), Institut du Cancer de Montpellier (ICM), Montpellier, France
| | - Diego Tosi
- Unité d'Essais de Phase Précoce (UEPP), Institut du Cancer de Montpellier (ICM), Montpellier, France - Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Montpellier, France
| |
Collapse
|
41
|
Understanding Inter-Individual Variability in Monoclonal Antibody Disposition. Antibodies (Basel) 2019; 8:antib8040056. [PMID: 31817205 PMCID: PMC6963779 DOI: 10.3390/antib8040056] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/22/2019] [Accepted: 11/27/2019] [Indexed: 12/29/2022] Open
Abstract
Monoclonal antibodies (mAbs) are currently the largest and most dominant class of therapeutic proteins. Inter-individual variability has been observed for several mAbs; however, an understanding of the underlying mechanisms and factors contributing to inter-subject differences in mAb disposition is still lacking. In this review, we analyze the mechanisms of antibody disposition and the putative mechanistic determinants of inter-individual variability. Results from in vitro, preclinical, and clinical studies were reviewed evaluate the role of the neonatal Fc receptor and Fc gamma receptors (expression and polymorphism), target properties (expression, shedding, turnover, internalization, heterogeneity, polymorphism), and the influence of anti-drug antibodies. Particular attention is given to the influence of co-administered drugs and disease, and to the physiological relevance of covariates identified by population pharmacokinetic modeling, as determinants of variability in mAb pharmacokinetics.
Collapse
|
42
|
Xia L, Liu W, Song Y, Zhu H, Duan Y. The Present and Future of Novel Protein Degradation Technology. Curr Top Med Chem 2019; 19:1784-1788. [PMID: 31644408 DOI: 10.2174/1568026619666191011162955] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/26/2019] [Accepted: 09/29/2019] [Indexed: 12/24/2022]
Abstract
Proteolysis targeting chimeras (PROTACs), as a novel therapeutic modality, play a vital role in drug discovery. Each PROTAC contains three key parts; a protein-of-interest (POI) ligand, a E3 ligase ligand, and a linker. These bifunctional molecules could mediate the degradation of POIs by hijacking the activity of E3 ubiquitin ligases for POI ubiquitination and subsequent degradation via the ubiquitin proteasome system (UPS). With several advantages over other therapeutic strategies, PROTACs have set off a new upsurge of drug discovery in recent years. ENDTAC, as the development of PROTACs technology, is now receiving more attention. In this review, we aim to summarize the rapid progress from 2018 to 2019 in protein degradation and analyze the challenges and future direction that need to be addressed in order to efficiently develop potent protein degradation technology.
Collapse
Affiliation(s)
- Liwen Xia
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, 450018, China.,State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Wei Liu
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, 450018, China
| | - Yinsen Song
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, 450018, China
| | - Hailiang Zhu
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, 450018, China
| | - Yongtao Duan
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, 450018, China
| |
Collapse
|
43
|
Yuan D, Rode F, Cao Y. A systems pharmacokinetic/pharmacodynamic model for concizumab to explore the potential of anti-TFPI recycling antibodies. Eur J Pharm Sci 2019; 138:105032. [PMID: 31394258 PMCID: PMC6824202 DOI: 10.1016/j.ejps.2019.105032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 07/22/2019] [Accepted: 08/01/2019] [Indexed: 01/31/2023]
Abstract
Concizumab is a humanized monoclonal antibody in clinical investigation directed against membrane-bound and soluble tissue factor pathway inhibitor (mTFPI and sTFPI) for treatment of hemophilia. Concizumab displays a non-linear pharmacokinetic (PK) profile due to mTFPI-mediated endocytosis and necessitates a high dose and frequent dosing to suppress the abundant sTFPI, a negative regulator of coagulation. Recycling antibodies that can dissociate bound mTFPI/sTFPI in endosomes for degradation and rescue antibody from degradation have a potential in reducing the dose by extending antibody systemic persistence and sTFPI suppression. We developed a systems PK/pharmacodynamics (PD) model with nested endosome compartments to simulate the effect of decreased antibody binding to mTFPI/sTFPI in endosomes on antibody clearance and sTFPI suppression for exploring the potential of anti-TFPI recycling antibodies in reducing the dose. A dynamic model-building strategy was taken. A reduced PK/PD model without the endosome compartments was developed to optimize unknown target turnover parameters using concizumab PK data. The optimized parameters were then employed in the systems PK/PD model for simulations. The obtained systems PK/PD model adequately described the PK of concizumab in rabbits, monkeys, and humans and the PD in humans. The systems PK/PD model predicted that an anti-TFPI recycling antibody with a 100-fold higher mTFPI/sTFPI dissociation constant in endosomes than concizumab can extend sTFPI suppression from 12 days to 1 month. Thus, the systems PK/PD model provides a quantitative platform for guiding the engineering and translational development of anti-TFPI recycling antibodies.
Collapse
Affiliation(s)
- Dongfen Yuan
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Frederik Rode
- Novo Nordisk, Translational DMPK, H. Lundbeck A/S, Denmark
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
44
|
Li D, Lee D, Dere RC, Zheng B, Yu S, Fuh FK, Kozak KR, Chung S, Bumbaca Yadav D, Nazzal D, Danilenko D, Go MAT, Williams M, Polson AG, Poon KA, Prabhu S. Evaluation and use of an anti-cynomolgus monkey CD79b surrogate antibody-drug conjugate to enable clinical development of polatuzumab vedotin. Br J Pharmacol 2019; 176:3805-3818. [PMID: 31270798 PMCID: PMC6780994 DOI: 10.1111/bph.14784] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 05/02/2019] [Accepted: 06/04/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE Polatuzumab vedotin is an antibody-drug conjugate (ADC) being developed for non-Hodgkin's lymphoma. It contains a humanized anti-CD79b IgG1 monoclonal antibody linked to monomethyl auristatin E (MMAE), an anti-mitotic agent. Polatuzumab vedotin binds to human CD79b only. Therefore, a surrogate ADC that binds to cynomolgus monkey CD79b was used to determine CD79b-mediated pharmacological effects in the monkey and to enable first-in-human clinical trials. EXPERIMENTAL APPROACH Polatuzumab vedotin, the surrogate ADC, and the corresponding antibodies were evaluated in different assays in vitro and in animals. In vitro assessments included binding to peripheral blood mononuclear cells from different species, binding to a human and monkey CD79b-expressing cell line, binding to human Fcγ receptors, and stability in plasma across species. In vivo, ADCs were assessed for anti-tumour activity in mice, pharmacokinetics/pharmacodynamics in monkeys, and toxicity in rats and monkeys. KEY RESULTS Polatuzumab vedotin and surrogate ADC bind with similar affinity to human and cynomolgus monkey B cells, respectively. Comparable in vitro plasma stability, in vivo anti-tumour activity, and mouse pharmacokinetics were also observed between the surrogate ADC and polatuzumab vedotin. In monkeys, only the surrogate ADC showed B-cell depletion and B-cell-mediated drug disposition, but both ADCs showed similar MMAE-driven myelotoxicity, as expected. CONCLUSIONS AND IMPLICATIONS The suitability of the surrogate ADC for evaluation of CD79b-dependent pharmacology was demonstrated, and anti-tumour activity, pharmacokinetics/pharmacodynamics, and toxicity data with both ADCs supported the entry of polatuzumab vedotin into clinical trials.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/immunology
- Antineoplastic Agents/pharmacology
- Binding Sites/drug effects
- Burkitt Lymphoma/drug therapy
- Burkitt Lymphoma/pathology
- CD79 Antigens/antagonists & inhibitors
- CD79 Antigens/immunology
- Cell Line
- Dose-Response Relationship, Drug
- Female
- Humans
- Immunoconjugates/chemistry
- Immunoconjugates/immunology
- Immunoconjugates/pharmacology
- Macaca fascicularis
- Male
- Mice
- Mice, SCID
- Molecular Conformation
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/pathology
- Rats
- Rats, Sprague-Dawley
- Receptors, IgG
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Dongwei Li
- Department of Pharmacokinetic and Pharmacodynamic SciencesGenentech, Inc.South San FranciscoCAUSA
| | - Donna Lee
- Department of Safety AssessmentGenentech, Inc.South San FranciscoCAUSA
| | - Randall C. Dere
- Department of BioAnalytical SciencesGenentech, Inc.South San FranciscoCAUSA
| | - Bing Zheng
- Department of Translational OncologyGenentech, Inc.South San FranciscoCAUSA
| | - Shang‐Fan Yu
- Department of Translational OncologyGenentech, Inc.South San FranciscoCAUSA
| | - Franklin K. Fuh
- Department of OMNI‐Biomarker DevelopmentGenentech, Inc.South San FranciscoCAUSA
| | - Katherine R. Kozak
- Department of Biochemical and Cellular PharmacologyGenentech, Inc.South San FranciscoCAUSA
| | - Shan Chung
- Department of BioAnalytical SciencesGenentech, Inc.South San FranciscoCAUSA
| | - Daniela Bumbaca Yadav
- Department of Pharmacokinetic and Pharmacodynamic SciencesGenentech, Inc.South San FranciscoCAUSA
| | - Denise Nazzal
- Department of BioAnalytical SciencesGenentech, Inc.South San FranciscoCAUSA
| | - Dimitry Danilenko
- Department of Safety AssessmentGenentech, Inc.South San FranciscoCAUSA
| | - Mary Ann T. Go
- Department of Translational OncologyGenentech, Inc.South San FranciscoCAUSA
| | - Marna Williams
- Department of Translational MedicineMedImmuneGaithersburgMDUSA
| | - Andrew G. Polson
- Department of Translational OncologyGenentech, Inc.South San FranciscoCAUSA
| | | | - Saileta Prabhu
- Department of Pharmacokinetic and Pharmacodynamic SciencesGenentech, Inc.South San FranciscoCAUSA
| |
Collapse
|
45
|
Kielbasa W, Helton DL. A new era for migraine: Pharmacokinetic and pharmacodynamic insights into monoclonal antibodies with a focus on galcanezumab, an anti-CGRP antibody. Cephalalgia 2019; 39:1284-1297. [PMID: 30917684 PMCID: PMC6710614 DOI: 10.1177/0333102419840780] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/17/2018] [Accepted: 03/04/2019] [Indexed: 12/15/2022]
Abstract
PURPOSE To review pharmacokinetic and pharmacodynamic characteristics of antibodies that bind to soluble ligands within the framework of calcitonin gene-related peptide antibodies. OVERVIEW Calcitonin gene-related peptide has been implicated in the pathophysiology of migraine. Galcanezumab is an antibody that binds to the ligand calcitonin gene-related peptide. Other antibodies that target calcitonin gene-related peptide include eptinezumab and fremanezumab. To understand how antibodies can affect the extent and duration of free ligand concentrations, it is important to consider the dose and pharmacokinetics of an antibody, and the kinetics of the ligand and antibody-ligand complex. Insights regarding the pharmacokinetic/pharmacodynamic properties of galcanezumab as a probe antibody drug and calcitonin gene-related peptide as its binding ligand regarding its clinical outcomes are provided. DISCUSSION Antibodies are administered parenterally because oral absorption is limited by gastrointestinal degradation and inefficient diffusion through the epithelium. The systemic absorption of antibodies following intramuscular or subcutaneous administration most likely occurs via convective transport through lymphatic vessels into blood. The majority of antibody elimination occurs via intracellular catabolism into peptides and amino acids following endocytosis. Binding of ligand to an antibody reduces the free ligand that is available to interact with the receptor and efficacy is driven by the magnitude and duration of the reduction in free ligand concentration. A galcanezumab pharmacokinetic/pharmacodynamic model shows that galcanezumab decreases free calcitonin gene-related peptide concentrations in a dose- and time-dependent manner and continues to suppress free calcitonin gene-related peptide with repeated dosing. The model provides evidence for a mechanistic linkage to galcanezumab therapeutic effects for the preventive treatment of migraine.
Collapse
|
46
|
Wu X, Nekka F, Li J. Analytical Solution and Exposure Analysis of a Pharmacokinetic Model with Simultaneous Elimination Pathways and Endogenous Production: The Case of Multiple Dosing Administration. Bull Math Biol 2019; 81:3436-3459. [PMID: 31420841 DOI: 10.1007/s11538-019-00651-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 07/15/2019] [Indexed: 12/17/2022]
Abstract
In this paper, a typical pharmacokinetic (PK) model is studied for the case of multiple intravenous bolus-dose administration. This model, of one-compartment structure, not only exhibits simultaneous first-order and Michaelis-Menten elimination, but also involves a constant endogenous production. For the PK characterization of the model, we have established the closed-form solution of concentrations over time, the existence and local stability of the steady state. Using analytical approaches and the concept of corrected concentration, we have shown that the area under the curve ([Formula: see text]) at steady state is higher compared to that at the single dose ([Formula: see text]). Moreover, by splitting the dose and dosing interval into halves, we have revealed that it can result in a significant decrease in the steady-state average concentration. These model-based findings, which contrast with the current knowledge for linear PK, confirm the necessity to revisit drugs exhibiting nonlinear PK and to suggest a rational way of using mathematical analysis for the dosing regimen design.
Collapse
Affiliation(s)
- Xiaotian Wu
- Department of Mathematics, Shanghai Maritime University, Shanghai, 201306, People's Republic of China
- Faculté de pharmacie, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Fahima Nekka
- Faculté de pharmacie, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Centre de Recherches Mathématiques, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Jun Li
- Faculté de pharmacie, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
- Centre de Recherches Mathématiques, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
| |
Collapse
|
47
|
D'Cunha R, Schmidt R, Widness JA, Mock DM, Yan X, Cress GA, Kuruvilla D, Veng-Pedersen P, An G. Target-mediated disposition population pharmacokinetics model of erythropoietin in premature neonates following multiple intravenous and subcutaneous dosing regimens. Eur J Pharm Sci 2019; 138:105013. [PMID: 31340188 DOI: 10.1016/j.ejps.2019.105013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/16/2019] [Accepted: 07/18/2019] [Indexed: 01/09/2023]
Abstract
Routine erythropoietin (Epo) therapy for neonatal anemia is presently controversial due to its modest response. We speculate that an important contributor to this modest response is that previous clinical study designs were not driven by rigorous mechanistic and kinetic insights into the complex pharmacokinetics (PK) and pharmacodynamics (PD) of Epo in this population. To address this therapeutic opportunity, we conducted a prospective clinical study to investigate the PK of Epo in very-low-birth-weight (VLBW) premature neonates using a unique Epo dosing algorithm that accounts for complex neonatal erythropoietic physiology. Twenty-seven subjects received up to 10 intravenous or subcutaneous exogenous doses of Epo (600 or 1200 U/kg) during the first 4 weeks of life. Subjects were administered two doses of Epo 1200 U/kg on days 2 and 16, and eight doses of Epo 600 U/kg on days 4, 5, 6, 7, 9, 14, 15, and 28 following birth. We have developed for the first time a mechanistic, target-mediated disposition model that provides novel insights into the mechanisms driving Epo PK in VLBW neonates. Epo association rate, kon, was estimated to be 0.00610 pM-1h-1, and the dissociation rate koff was 0.112 h-1. Internalization of the Epo-target complex (kint) and the total receptor concentration (Rmax) were estimated to be 0.118 h-1 and 133 pM, respectively. Following s.c. administration, the absorption rate (ka) of Epo was 0.0738h-1 and bioavailability was 78.0%. Our mechanism-based population pharmacokinetic analysis provided quantitative insight into Epo kinetics in VLBW neonates; the information gained will assist in deriving dosing strategies for neonatal anemia and for neuroprotection efficacy studies.
Collapse
Affiliation(s)
- Ronilda D'Cunha
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Robert Schmidt
- Department of Pediatrics Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - John A Widness
- Department of Pediatrics Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Donald M Mock
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, USA
| | - Xiaoyu Yan
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong
| | - Gretchen A Cress
- Department of Pediatrics Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Denison Kuruvilla
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA; MedImmune, LLC., San Francisco, CA, USA
| | - Peter Veng-Pedersen
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Guohua An
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
48
|
Bartelink IH, Jones EF, Shahidi‐Latham SK, Lee PRE, Zheng Y, Vicini P, van ‘t Veer L, Wolf D, Iagaru A, Kroetz DL, Prideaux B, Cilliers C, Thurber GM, Wimana Z, Gebhart G. Tumor Drug Penetration Measurements Could Be the Neglected Piece of the Personalized Cancer Treatment Puzzle. Clin Pharmacol Ther 2019; 106:148-163. [PMID: 30107040 PMCID: PMC6617978 DOI: 10.1002/cpt.1211] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/30/2018] [Indexed: 12/30/2022]
Abstract
Precision medicine aims to use patient genomic, epigenomic, specific drug dose, and other data to define disease patterns that may potentially lead to an improved treatment outcome. Personalized dosing regimens based on tumor drug penetration can play a critical role in this approach. State-of-the-art techniques to measure tumor drug penetration focus on systemic exposure, tissue penetration, cellular or molecular engagement, and expression of pharmacological activity. Using in silico methods, this information can be integrated to bridge the gap between the therapeutic regimen and the pharmacological link with clinical outcome. These methodologies are described, and challenges ahead are discussed. Supported by many examples, this review shows how the combination of these techniques provides enhanced patient-specific information on drug accessibility at the tumor tissue level, target binding, and downstream pharmacology. Our vision of how to apply tumor drug penetration measurements offers a roadmap for the clinical implementation of precision dosing.
Collapse
Affiliation(s)
- Imke H. Bartelink
- Department of MedicineUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Clinical Pharmacology, Pharmacometrics and DMPK (CPD)MedImmuneSouth San FranciscoCaliforniaUSA
- Department of Clinical Pharmacology and PharmacyAmsterdam UMCVrije Universiteit AmsterdamThe Netherlands
| | - Ella F. Jones
- Department of Radiology and Biomedical ImagingUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | | | - Pei Rong Evelyn Lee
- Department of Laboratory Medicine of the UCSF Helen Diller Family Comprehensive Cancer CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Yanan Zheng
- Clinical Pharmacology, Pharmacometrics and DMPK (CPD)MedImmuneSouth San FranciscoCaliforniaUSA
| | - Paolo Vicini
- Clinical Pharmacology, Pharmacometrics and DMPK (CPD)MedImmuneCambridgeUK
| | - Laura van ‘t Veer
- Department of Laboratory Medicine of the UCSF Helen Diller Family Comprehensive Cancer CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Denise Wolf
- Department of Laboratory Medicine of the UCSF Helen Diller Family Comprehensive Cancer CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging at Stanford Health CareStanfordCaliforniaUSA
| | - Deanna L. Kroetz
- Department of Bioengineering and Therapeutic Sciences (BTS)School of PharmacyUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Brendan Prideaux
- Rutgers New Jersey Medical SchoolPublic Health Research InstituteRutgers, The State University of New JerseyNew BrunswickNew JerseyUSA
| | - Cornelius Cilliers
- Departments of Chemical Engineering and Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Greg M. Thurber
- Departments of Chemical Engineering and Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Zena Wimana
- Institut Jules BordetUniversité Libre de Bruxelles (ULB)BrusselsBelgium
| | - Geraldine Gebhart
- Institut Jules BordetUniversité Libre de Bruxelles (ULB)BrusselsBelgium
| |
Collapse
|
49
|
Tang H, Liu Y, Yu Z, Sun M, Lin L, Liu W, Han Q, Wei M, Jin Y. The Analysis of Key Factors Related to ADCs Structural Design. Front Pharmacol 2019; 10:373. [PMID: 31068807 PMCID: PMC6491742 DOI: 10.3389/fphar.2019.00373] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/26/2019] [Indexed: 01/27/2023] Open
Abstract
Antibody–drug conjugates (ADCs) have developed rapidly in recent decades. However, it is complicated to map out a perfect ADC that requires optimization of multiple parameters including antigens, antibodies, linkers, payloads, and the payload-linker linkage. The therapeutic targets of the ADCs are expected to express only on the surface of the corresponding target tumor cells. On the contrary, many antigens usually express on normal tissues to some extent, which could disturb the specificity of ADCs and limit their clinical application, not to mention the antibody is also difficult to choose. It requires to not only target and have affinity with the corresponding antigen, but it also needs to have a linkage site with the linker to load the payloads. In addition, the linker and payload are indispensable in the efficacy of ADCs. The linker is required to stabilize the ADC in the circulatory system and is brittle to release free payload while the antibody combines with antigen. Also, it is a premise that the dose of ADCs will not kill normal tissues and the released payloads are able to fulfill the killing potency in tumor cells at the same time. In this review, we mainly focus on the latest development of key factors affecting ADCs progress, including the selection of antibodies and antigens, the optimization of payload, the modification of linker, payload-linker linkage, and some other relevant parameters of ADCs.
Collapse
Affiliation(s)
- Haichao Tang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, China
| | - Yan Liu
- Liaoning Research Institute of Family Planning, Shenyang, China
| | - Zhaojin Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, China
| | - Mingli Sun
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, China
| | - Lu Lin
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, China
| | - Wensi Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, China
| | - Qiang Han
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, China
| | - Ying Jin
- Liaoning Research Institute of Family Planning, Shenyang, China
| |
Collapse
|
50
|
Iyengar ARS, Gupta S, Jawalekar S, Pande AH. Protein Chimerization: A New Frontier for Engineering Protein Therapeutics with Improved Pharmacokinetics. J Pharmacol Exp Ther 2019; 370:703-714. [DOI: 10.1124/jpet.119.257063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/16/2019] [Indexed: 12/20/2022] Open
|